US20040265269A1 - Cysteine variants of granulocyte colony-stimulating factor - Google Patents

Cysteine variants of granulocyte colony-stimulating factor Download PDF

Info

Publication number
US20040265269A1
US20040265269A1 US10/856,219 US85621904A US2004265269A1 US 20040265269 A1 US20040265269 A1 US 20040265269A1 US 85621904 A US85621904 A US 85621904A US 2004265269 A1 US2004265269 A1 US 2004265269A1
Authority
US
United States
Prior art keywords
cysteine
leu
substituted
ser
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US10/856,219
Other versions
US7309781B2 (en
Inventor
George Cox
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bolder Biotechnology Inc
Original Assignee
Bolder Biotechnology Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bolder Biotechnology Inc filed Critical Bolder Biotechnology Inc
Priority to US10/856,219 priority Critical patent/US7309781B2/en
Publication of US20040265269A1 publication Critical patent/US20040265269A1/en
Application granted granted Critical
Publication of US7309781B2 publication Critical patent/US7309781B2/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: BOLDER BIOTECHNOLOGY, INC.
Adjusted expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5431IL-11
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/10Drugs for disorders of the endocrine system of the posterior pituitary hormones, e.g. oxytocin, ADH
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/505Erythropoietin [EPO]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/53Colony-stimulating factor [CSF]
    • C07K14/535Granulocyte CSF; Granulocyte-macrophage CSF
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5403IL-3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5406IL-4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5409IL-5
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5412IL-6
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5418IL-7
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5425IL-9
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5428IL-10
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5434IL-12
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5437IL-13
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5443IL-15
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/55IL-2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/565IFN-beta
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/57IFN-gamma
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/61Growth hormones [GH] (Somatotropin)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to genetically engineered therapeutic proteins. More specifically, the engineered proteins include growth hormone and related proteins.
  • GH growth hormone
  • EPO erythropoietin
  • TPO thrombopoietin
  • IL-2 interleukin-2
  • IL-3 IL-4, IL-5, IL-6, IL-7, IL-9, IL-10, IL-11, IL-12 (p35 subunit) IL-13, IL-15
  • oncostatin M ciliary neurotrophic factor, leukemia inhibitory factor, alpha interferon, beta interferon, gamma interferon, omega interferon, tau interferon, granulocyte-colony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating factor (M-CSF) and cardiotrophin-1
  • the length of time an injected protein remains in the body is finite and is determined by, e.g., the protein's size and whether or not the protein contains covalent modifications such as glycosylation. Circulating concentrations of injected proteins change constantly, often by several orders of magnitude, over a 24-hour period. Rapidly changing concentrations of protein agonists can have dramatic downstream consequences, at times under-stimulating and at other times over-stimulating target cells. Similar problems plague protein antagonists. These fluctuations can lead to decreased efficacy and increased frequency of adverse side effects for protein therapeutics. The rapid clearance of recombinant proteins from the body significantly increases the amount of protein required per patient and dramatically increases the cost of treatment. The cost of human protein pharmaceuticals is expected to increase dramatically in the years ahead as new and existing drugs are approved for more disease indications.
  • the present invention provides a solution to this problem by providing methods to prolong the circulating half-lives of protein therapeutics in the body so that the proteins do not have to be injected frequently. This solution also satisfies the needs and desires of patients for protein therapeutics that are “user-friendly”, i.e., protein therapeutics that do not require frequent injections.
  • the present invention solves these and other problems by providing biologically active, cysteine-added variants of members of the growth hormone supergene family.
  • the invention also provides for the chemical modification of these variants with cysteine-reactive polymers or other types of cysteine-reactive moieties to produce derivatives thereof and the molecules so produced.
  • the present invention provides cysteine variants of members of the GH supergene family.
  • the variants comprise a cysteine residue substituted for a nonessential amino acid of the proteins.
  • the variants comprise a cysteine residue substituted for an amino acid selected from amino acids in the loop regions, the ends of the alpha helices, proximal to the first amphipathic helix, and distal to the final amphipathic helix or wherein the cysteine residue is added at the N-terminus or C-terminus of the proteins.
  • Preferred sites for substitution are the N— and O-linked glycosylation sites.
  • cysteine variants wherein the amino acid substituted for is in the A-B loop, B-C loop, the C-D loop or D-E loop of interferon/interferon-10-like members of the GH supergene family.
  • cysteine variants of members of the GH supergene family wherein the cysteine residue is introduced between two amino acids in the natural protein.
  • the cysteine residue is introduced into the loop regions, the ends of the alpha helices, proximal to the first amphipathic helix, or distal to the final amphipathic helix.
  • the cysteine variant is introduced between two amino acids in an N—O-linked glycosylation site or adjacent to an amino acid in an N-linked or O-linked glycosylation site.
  • cysteine variants wherein the loop region where the cysteine is introduced is the A-B loop, the B-C loop, the C-D loop or D-E loop of interferon/interferon-10-like members of the GH supergene family.
  • cysteine substitutions or insertion mutations also can include the insertion of one or more additional amino acids amino acids at the amino-terminal or carboxy-terminal to the cysteine substitution or insertion.
  • cysteine variants that are further derivatised by PEGylating the cysteine variants and including the derivatised proteins produced thereby.
  • cysteine variants of the members of the GH supergene family also are provided, including for example, variants of GH.
  • the GH cysteine variants can have the substituted-for amino acid or inserted cysteine located at the N-terminal end of the A-B loop, the B-C loop, the C-D loop, the first three or last three amino acids in the A, B, C and D helices and the amino acids proximal to helix A and distal to helix D.
  • cysteine can be substituted for the following amino acids: F1, T3, P5, E33, A34, K38, E39, Q40, S43, Q46, N47, P48, Q49, T50, S51, S55, T60, A98, N99, S100, G104, A105, S106, E129, D130, G131, S132, P133, T135, G136, Q137, K140, Q141, T142, S144, K145, D147, T148, N149, S150, H151, N152, D153, S184, E186, G187, S188, and G190.
  • cysteine variants according to the invention include erythropoietin variants.
  • Erythropoietin variants include those wherein the substituted for amino acid is located in the A-B loop, the B-C loop, the C-D loop, the amino acids proximal to helix A and distal to helix D and the N— or C-terminus.
  • the EPO cysteine variants include molecules wherein the amino acids indicated below have a cysteine substituted therefor: serine-126, N24, I25, T26, N38, I39, T40, N83, S84, A1, P2, P3, R4, D8, S9, T27, G28, A30, E31, H32, S34, N36, D43, T44, K45, N47, A50, K52, E55, G57, Q58, G77, Q78, A79, Q86, W88, E89, T107, R110, A111, G113, A114, Q15, K116, E117, A118, S120, P121, P122, D123, A124, A125, A127, A128, T132, K154, T157, G158, E159, A160, T163, G164, D165, R166 and S85.
  • the members of the GH supergene family include growth hormone, prolactin, placental lactogen, erythropoietin, thrombopoietin, interleukin-2, interleukin-3, interleukin-4, interleukin-5, interleukin-6, interleukin-7, interleukin-9, interleukin-10, interleukin-1, interleukin-12 (p35 subunit), interleukin-13, interleukin-15, oncostatin M, ciliary neurotrophic factor, leukemia inhibitory factor, alpha interferon, beta interferon, gamma interferon, omega interferon, tau interferon, granulocyte-colony stimulating factor, granulocyte-macrophage colony stimulating factor, macrophage colony stimulating factor, cardiotrophin-1 and other proteins identified and classified as members of the family.
  • the proteins can be derived from any animal species including human, companion animals and farm animals.
  • the present invention relates to cysteine variants and, among other things, the site-specific conjugation of such proteins with polyethylene glycol (PEG) or other such moieties.
  • PEG polyethylene glycol
  • PEG is a non-antigenic, inert polymer that significantly prolongs the length of time a protein circulates in the body. This allows the protein to be effective for a longer period of time.
  • Covalent modification of proteins with PEG has proven to be a useful method to extend the circulating half-lives of proteins in the body (Abuchowski et al., 1984; Hershfield, 1987; Meyers et al., 1991). Covalent attachment of PEG to a protein increases the protein's effective size and reduces its rate of clearance rate from the body.
  • PEGs are commercially available in several sizes, allowing the circulating half-lives of PEG-modified proteins to be tailored for individual indications through use of different size PEGs.
  • Other benefits of PEG modification include an increase in protein solubility, an increase in in vivo protein stability and a decrease in protein immunogenicity (Katre et al., 1987; Katre, 1990).
  • the preferred method for PEGylating proteins is to covalently attach PEG to cysteine residues using cysteine-reactive PEGs.
  • cysteine-reactive PEGs A number of highly specific, cysteine-reactive PEGs with different reactive groups (e.g., maleimide, vinylsulfone) and different size PEGs (2-20 kDa) are commercially available (e.g., from Shearwater, Polymers, Inc., Huntsville, Ala.).
  • these PEG reagents selectively attach to “free” cysteine residues, i.e., cysieine residues not involved in disulfide bonds.
  • the conjugates are hydrolytically stable. Use of cysteine-reactive PEGs allows the development of homogeneous PEG-protein conjugates of defined structure.
  • the newly added “free” cysteines can serve as sites for the specific attachment of a PEG molecule using cysteine-reactive PEGs.
  • the added cysteine must be exposed on the protein's surface and accessible for PEGylation for this method to be successful. If the site used to introduce an added cysteine site is non-essential for biological activity, then the PEGylated protein will display essentially wild type (normal) in vitro bioactivity.
  • the major technical challenge in PEGylating proteins with cysteine-reactive PEGs is the identification of surface exposed, non-essential regions in the target protein where cysteine residues can be added or substituted for existing amino acids without loss of bioactivity.
  • cysteine-added variants of a few human proteins and PEG-polymer conjugates of these proteins have been described.
  • U.S. Pat. No. 5,206,344 describes cysteine-added variants of IL-2. These cysteine-added variants are located within the first 20 amino acids from the amino terminus of the mature IL-2 polypeptide chain. The preferred cysteine variant is at position 3 of the mature polypeptide chain, which corresponds to a threonine residue that is O-glycosylated in the naturally occurring protein.
  • U.S. Pat. No. 5,166,322 teaches cysteine added variants of IL-3. This variants are located within the first 14 amino acids from the N-terminus of the mature protein sequence. The patent teaches expression of the proteins in bacteria and covalent modification of the proteins with cysteine-reactive PEGs. No information is provided as to whether the cysteine-added variants and PEG-conjugates of IL-3 are biologically active. Cysteine-added variants at other positions in the polypeptide chain were not reported.
  • IGF-I insulin-like growth factor-I
  • IGF-I insulin-like growth factor-I
  • World patent application WO9422466 teaches two cysteine-added variants of insulin-like growth factor (IGF) binding protein-1, which has a very different structure than GH and is not a member of the GH supergene family.
  • the two cysteine-added IGF binding protein-1 variants disclosed are located at positions 98 and 101 in the mature protein chain and correspond to serine residues that are phosphorylated in the naturally-occurring protein.
  • U.S. patent application Ser. No. 07/822296 teaches cysteine added variants of tumor necrosis factor binding protein, which is a soluble, truncated form of the tumor necrosis factor cellular receptor.
  • Tumor necrosis factor binding protein has a very different structure than GH and is not a member of the GH supergene family.
  • IGF-I, IGF binding protein-1 and tumor necrosis factor binding protein have secondary and tertiary structures that are very different from GH and the proteins are not members of the GH supergene family. Because of this, it is difficult to use the information gained from studies of IGF-I, IGF binding protein-l and tumor necrosis factor binding protein to create cysteine-added variants of members of the GH supergene family.
  • the studies with IL-2 and IL-3 were carried out before the structures of IL-2 and IL-3 were known (McKay 1992; Bazan, 1992) and before it was known that these proteins are members of the GH supergene family. Previous experiments aimed at identifying preferred sites for adding cysteine residues to IL-2 and IL-3 were largely empirical and were performed prior to experiments indicating that members of the GH supergene family possessed similar secondary and tertiary structures.
  • the present invention Based on the structural information now available for members of the GH supergene family, the present invention provides “rules” for determining a priori which regions and amino acid residues in members of the GH supergene family can be used to introduce or substitute cysteine residues without significant loss of biological activity.
  • these cysteine-added variants of members of the GH supergene family will possess novel properties such as the ability to be covalently modified at defined sites within the polypeptide chain with cysteine-reactive polymers or other types of cysteine-reactive moieties.
  • the covalently modified proteins will be biologically active.
  • GH is the best-studied member of the GH supergene family.
  • GH is a 22 kDa protein secreted by the pituitary gland.
  • GH stimulates metabolism of bone, cartilage and muscle and is the body's primary hormone for stimulating somatic growth during childhood.
  • Recombinant human GH (rhGH) is used to treat short stature resulting from GH inadequacy and renal failure in children.
  • GH is not glycosylated and can be produced in a fully active form in bacteria.
  • the protein has a short in vivo half-life and must be administered by daily subcutaneous injection for maximum effectiveness (MacGillivray et al., 1996).
  • Recombinant human GH (rhGH) was approved recently for treating cachexia in AIDS patients and is under study for treating cachexia associated with other diseases.
  • GH The sequence of human GH is well known (see, e.g., Martial et al. 1979; Goeddel et al. 1979 which are incorporated herein by reference; SEQ ID NO:1). GH is closely related in sequence to prolactin and placental lactogen and these three proteins were considered originally to comprise a small gene family. The primary sequence of GH is highly conserved among animal species (Abdel-Meguid et al., 1987), consistent with the protein's broad species cross-reactivity. The three dimensional folding pattern of porcine GH has been solved by X-ray crystallography (Abdel-Meguid et al., 1987).
  • the protein has a compact globular structure, comprising four amphipathic alpha helical bundles joined by loops.
  • Human GH has a similar structure (de Vos et al., 1992).
  • the four alpha helical regions are termed A-D beginning from the N-terminus of the protein.
  • the loop regions are referred to by the helical regions they join, e.g., the A-B loop joins helical bundles A and B.
  • the A-B and C-D loops are long, whereas the B-C loop is short.
  • GH contains four cysteine residues, all of which participate in disulfide bonds.
  • the disuifide assignments are cysteine53 joined to cysteine165 and cysteine182 joined to cysteine189.
  • site I encompasses the Carboxy (C)-terminal end of helix D and parts of helix A and the A-B loop
  • site II encompasses the Amino (N)-terminal region of helix A and a portion of helix C. Binding of GH to its receptor occurs sequentially, with site I always binding first.
  • Site II then engages a second GH receptor, resulting in receptor dimerization and activation of the intracellular signaling pathways that lead to cellular responses to GH.
  • a GH mutein in which site II has been mutated (a glycine to arginine mutation at amino acid 120) is able to bind a single GH receptor, but is unable to dimerize GH receptors; this mutein acts as a GH antagonist in vitro, presumably by occupying GH receptor sites without activating intracellular signaling pathways (Fuh et al., 1992).
  • Alanine scanning mutagenesis (replacement of individual amino acids with alanine) identified 14 amino acids that are critical for GH bioactivity (Cunningham and Wells, 1989). These amino acids are located in the helices A, B, C, and D and the A-B loop and correspond to sites I and II identified from the structural studies. Two lysine residues at amino acid positions 41 and 172, K41 and K172, were determined to be critical components of the site I receptor binding site, which explains the decrease in bioactivity observed when K1 72 is acetylated (Teh and Chapman, 1988).
  • cytokines including G-CSF (Hill et al., 1993), GM-CSF (Diederichs et al., 1991; Walter et al., 1992), IL-2 (Bazan, 1992; McKay, 1992), IL-4 ( Redfield et al., 1991; Powers et al., 1992), and IL-5 (Milburn et al., 1993) have been determined by X-ray diffraction and NMR studies and show striking conservation with the GH structure, despite a lack of significant primary sequence homology.
  • EPO is considered to be a member of this family based upon modeling and mutagenesis studies (Boissel et al., 1993; Wen et al., 1994).
  • a large number of additional cytokines and growth factors including ciliary neurotrophic factor (CNTF), leukemia inhibitory factor (LIEF), thrombopoietin (TPO), oncostatin M, macrophage colony stimulating factor (M-CSF), IL-3, IL-6, IL-7, IL-9, IL-12, IL-13, IL-15, and alpha, beta, omega, tau and gamma interferon belong to this family (reviewed in Mott and Campbell, 1995; Silvennoinen and Ihle 1996). All of the above cytokines and growth factors are now considered to comprise one large gene family, of which GH is the prototype.
  • GH family members In addition to sharing similar secondary and tertiary structures, members of this family share the property that they must oligomerize cell surface receptors to activate intracellular signaling pathways.
  • Some GH family members e.g., GH and EPO, bind a single type of receptor and cause it to form homodimers.
  • Other family members e.g., IL-2, IL-4, and IL-6, bind more than one type of receptor and cause the receptors to form heterodimers or higher order aggregates (Davis et al., 1993; Paonessa et al., 1995; Mott and Campbell, 1995).
  • a general conclusion reached from mutational studies of various members of the GH supergene family is that the loops joining the alpha helices generally tend to not be involved in receptor binding.
  • the short B-C loop appears to be non-essential for receptor binding in most, if not all, family members.
  • the B-C loop is a preferred region for introducing cysteine substitutions in members of the GH supergene family.
  • the A-B loop, the B-C loop, the C-D loop (and D-E loop of interferon/IL-10-like members of the GH superfamily) also are preferred sites for introducing cysteine mutations.
  • Amino acids proximal to helix A and distal to the final helix also tend not to be involved in receptor binding and also are preferred sites for introducing cysteine substitutions.
  • Certain members of the GH family e.g., EPO, IL-2, IL-3, IL-4, IL-6, G-CSF, GM-CSF, TPO, IL-10, IL-12 p35, IL-13, IL-15 and beta-interferon contain N-linked and O-linked sugars. The glycosylation sites in the proteins occur almost exclusively in the loop regions and not in the alpha helical bundles.
  • the loop regions generally are not involved in receptor binding and because they are sites for the covalent attachment of sugar groups, they are preferred sites for introducing cysteine substitutions into the proteins.
  • Amino acids that comprise the N— and O-linked glycosylation sites in the proteins are preferred sites for cysteine substitutions because these amino acids are surface-exposed, the natural protein can tolerate bulky sugar groups attached to the proteins at these sites and the glycosylation sites tend to be located away from the receptor binding sites.
  • GH gene family Many additional members of the GH gene family are likely to be discovered in the future. New members of the GH supergene family can be identified through computer-aided secondary and tertiary structure analyses of the predicted protein sequences. Members of the GH supergene family will possess four or five amphipathic helices joined by non-helical amino acids (the loop regions). The proteins may contain a hydrophobic signal sequence at their N-terminus to promote secretion from the cell. Such later discovered members of the GH supergen family also are included within this invention.
  • the present invention provides “rules” for creating biologically active cysteine-added variants of members of the GH supergene family. These “rules” can be applied to any existing or future member of the GH supergene family.
  • the cysteine-added variants will posses novel properties not shared by the naturally occurring proteins.
  • the cysteine added variants will possess the property that they can be covalently modified with cysteine-reactive polymers or other types of cysteine-reactive moieties to generate biologically active proteins with improved properties such as increased in vivo half-life, increased solubility and improved in vivo efficacy.
  • the present invention provides biologically active cysteine variants of members of the GH supergene family by substituting cysteine residues for non-essential amino acids in the proteins.
  • the cysteine residues are substituted for amino acids that comprise the loop regions, for amino acids near the ends of the alpha helices and for amino acids proximal to the first amphipathic helix or distal to the final amphipathic helix of these proteins.
  • Other preferred sites for adding cysteine residues are at the N-terminus or C-terminus of the proteins.
  • Cysteine residues also can be introduced between two amino acids in the disclosed regions of the polypeptide chain.
  • N— and O-linked glycosylation sites in the proteins are preferred sites for introducing cysteine substitutions either by substitution for amino acids that make up the sites or, in the case of N-linked sites, introduction of cysteines therein.
  • the glycosylation sites can be serine or threonine residues that are O-glycosylated or asparagine residues that are N-glycosylated.
  • N-linked glycosylation sites have the general structure asparagine-X-serine or threonine (N—X—S/T), where X can be any amino acid.
  • the asparagine residue, the amino acid in the X position and the serine/threonine residue of the N-linked glycosylation site are preferred sites for creating biologically active cysteine-added variants of these proteins.
  • Amino acids immediately surrounding or adjacent to the O-linked and N-linked glycosylation sites are preferred sites for introducing cysteine-substitutions.
  • certain of the “rules” for identifying preferred sites for creating biologically active cysteine-added protein variants can be applied to any protein, not just proteins that are members of the GH supergene family.
  • preferred sites for creating biologically active cysteine variants of proteins are O-linked glycosylation sites. Amino acids immediately surrounding the O-linked glycosylation site (within about 10 residues on either side of the glycosylation site) also are preferred sites.
  • N-linked glycosylation sites, and the amino acid residues immediately adjacent on either side of the glycosylation site also are preferred sites for creating cysteine added protein variants.
  • cysteine-added protein variants Amino acids that can be replaced with cysteine without significant loss of biological activity also are preferred sites for creating cysteine-added protein variants.
  • Such non-essential amino acids can be identified by performing cysteine-scanning mutagenesis on the target protein and measuring effects on biological activity. Cysteine-scanning mutagenesis entails adding or substituting cysteine residues for individual amino acids in the polypeptide chain and determining the effect of the cysteine substitution on biological activity. Cysteine scanning mutagenesis is similar to alanine-scanning mutagenesis (Cunningham et al., 1992), except that target amino acids are individually replaced with cysteine rather than alanine residues.
  • One strategy involves engineering the cytokines and growth factors so that they can bind to, but not oligomerize receptors. This is accomplished by mutagenizing the second receptor binding site (site II) on the molecules. The resulting muteins are able to bind and occupy receptor sites but are incapable of activating intracellular signaling pathways. This strategy has been successfully applied to GH to make a GH antagonist (Cunningham et al., 1992).
  • Cunningham et al. (1992) developed an in vitro GH antagonist by mutating a glycine residue (amino acid 120) to an arginine.
  • This glycine residue is a critical component of the second receptor binding site in GH; when it is replaced with arginine, GH cannot dimerize receptors.
  • the glycine to arginine mutation at position 120 can be introduced into DNA sequences encoding the cysteine-added variants of GH contemplated herein to create a cysteine-added GH antagonist that can be conjugated with cysteine-reactive PEGs or other types of cysteine-reactive moieties.
  • amino acid changes in other proteins that turn the proteins from agonists to antagonists could be incorporated into DNA sequences encoding cysteine-added protein variants described herein.
  • Hercus et al.(1994) reported that substituting arginine or lysine for glutamic acid at position 21 in the mature GM-CSF protein converts GM-CSF from an agonist to an antagonist.
  • Tavernier et al.(1995) reported that substituting glutamine for glutamic acid at position 13 of mature IL-5 creates an IL-5 antagonist.
  • Proteins that are members of the GH supergene family are provided in Silvennoimem and Ihle (1996).
  • Silvennoimem and lhle (1996) also provide information about the structure and expression of these proteins.
  • DNA sequences, encoded amino acids and in vitro and in vivo bioassays for the proteins described herein are described in Aggarwal and Gutterman (1992; 1996), Aggarwal (1998), and Silvennoimem and lhle (1996). Bioassays for the proteins also are provided in catalogues of various commercial suppliers of these proteins such as R&D Systems, Inc. and Endogen, Inc.
  • This example discloses certain amino acids in GH that are non-essential for biological activity and which, when mutated to cysteine residues, will not alter the normal disulfide binding pattern and overall conformation of the molecule. These amino acids are located at the N-terminal end of the A-B loop (amino acids 34-52 of the mature protein sequence; SEQ ID NO: 1; Martial et al 1979; Goeddel et al 1979), the B-C loop (amino acids 97-105 in the mature protein sequence), and the C-D loop (amino acids 130-153 in the mature protein sequence). Also identified as preferred sites for introducing cysteine residues are the first three or last three amino acids in the A, B, C and D helices and the amino acids proximal to helix A and distal to helix D.
  • DNA sequences encoding wild type GH can be amplified using the polymerase chain reaction technique from commercially available single-stranded cDNA prepared from human pituitaries (ClonTech, San Diego, Calif.) or assembled using overlapping oligonucleotides. Specific mutations can be introduced into the GH sequence using a variety of procedures such as phage techniques (Kunkel et al 1987), PCR mutagenesis techniques (Innis et al 1990; White 1993) mutagenesis kits such as those sold by Stratagene (“Quick-Change Mutagenesis” kit, San Diego, Calif.) or Promega (Gene Editor Kit, Madison Wis.).
  • Cysteine substitutions can be introduced into any of the amino acids comprising the B-C loop, C-D loop and N-terminal end of the A-B loop or into the first three amino acids of the alpha helical regions that adjoin these regions or in the region proximal to helix A or distal to helix D.
  • Preferred sites for introduction of cysteine residues are: F1, T3, P5, E33, A34, K38, E39, Q40, S43, Q46, N47, P48, Q49, T50, S51, S55, T60, A98, N99, S100, G104, A105, S106, E129, D130, G131, S132, P133, T135, G136, Q137, K140, Q141, T142, S144, K145, D147, T148, N149, S150, H151, N152, D153, S184, E186, G187, S188, and G190.
  • Cysteine residues also can be introduced at the beginning of the mature protein, i.e., proximal to the F1 amino acid, or following the last amino acid in the mature protein, i.e., following F191. If desirable, two or more such mutations can be readily combined in the same protein either by in vitro DNA recombination of cloned mutant genes and/or sequential construction of individual desired mutations.
  • the human GH gene was amplified from human pituitary single-stranded cDNA (commercially available from CLONTECH, Inc., Palo Alto, Calif.) using the polymerase chain reaction (PCR) technique and primers BB1 and BB2.
  • the sequence of BB1 is 5′-GGGGGTCGACCATATGTTCCCAACCATTCCCTTATCCAG-3′ (SEQ ID NO: 24).
  • the sequence of BB2 is 5′-GGGGGATCCTCACTAGAAGCCACAGCTGCCCTC-3′ (SEQ ID NO: 25).
  • Primer BB1 was designed to encode an initiator methionine preceding the first amino acid of mature GH, phenylalanine, and SalI and NdeI sites for cloning purposes.
  • the reverse primer, BB2 contains a BamHI site for cloning purposes.
  • the PCR 100 microliter reactions contained 20 pmoles of each oligonucleotide primer, 1 ⁇ PCR buffer (Perkin-Elmer buffer containing MgCl 2 ), 200 micromolar concentration of each of the four nucleotides dA, dC, dG and dT, 2 ng of single-stranded cDNA, 2.5 units of Taq polymerase (Perkin-Elmer) and 2.5 units of Pfu polymerase (Stratagene, Inc).
  • the PCR reaction conditions were 96° C. for 3 minutes, 35 cycles of (95° C., 1 minute; 63° C. for 30 seconds; 72° C.
  • thermocycler employed was the Amplitron II Thermal Cycler (Thermolyne).
  • the approximate 600 bp PCR product was digested with SalI and BamHI, gel purified and cloned into similarly digested plasmid pUC19 (commercially available from New England BioLabs, Beverly, Mass.).
  • the ligation mixture was transformed into E. coli strain DH5alpha and transformants selected on LB plates containing ampicillin. Several colonies were grown overnight in LB media and plasmid DNA isolated using miniplasmid DNA isolation kits purchased from Qiagen, Inc (Valencia, Calif.). Clone LB6 was determined to have the correct DNA sequence.
  • clone LB6 was digested with NdeI and EcoRI, the approximate 600 bp fragment gel-purified, and cloned into plasmid pCYB1 (commercially available from New England BioLabs, Beverly, Mass.) that had been digested with the same enzymes and phosphatased.
  • the ligation mixture was transformed into E. coli DH5alpha and transformants selected on LB ampicillin plates. Plasmid DNA was isolated from several transformants and screened by digestion with NdeI and EcoRI. A correct clone was identified and named pCYB1: wtGH (pBBT120). This plasmid was transformed into E. coli strains JM109 or W3110 (available from New England BioLabs and the American Type Culture Collection).
  • Wild type GH clone LB6 (pUC19: wild type GH) was used as the template to construct a GH clone containing the E. coli STII signal sequence (Picken et al. 1983). Because of its length, the STII sequence was added in two sequential PCR reactions. The first reaction used forward primer BB12 and reverse primer BB10. BB10 has the sequence: (SEQ ID NO: 28) 5′CGCGGATCCGATTAGAATCCACAGCTCCCCTC3′.
  • BB12 has the sequence: 5′ATCTATGTTCGTTTTCTCTATCGCTACCAACGCTTACGCATTCCCAACCATTCCCTTATCCAG-3′. (SEQ ID NO: 30)
  • PCR reactions were as described for amplifying wild type GH except that approximately 4 ng of plasmid LB6 was used as the template rather than single-stranded cDNA and the PCR conditions were 96° C. for 3 minutes, 30 cycles of (95° C. for 1 minute; 63° C. for 30 seconds; 72° C. for 1 minute) followed by 72° C. for 10 minutes.
  • the approximate 630 bp PCR product was gel-purified using the Qiaex II Gel Extraction Kit (Qiagen, Inc), diluted 50-fold in water and 2 microliters used as template for the second PCR reaction.
  • the second PCR reaction used reverse primer BB10 and forward primer BB11.
  • BB11 has the sequence: 5′CCCCCTCTAGACATATGAAGAAGAACATCGCATTCCTGCTGGCATCTATGTTCGTTTTCTCTATCG-3′. (SEQ ID NO: 29)
  • Primer BB11 contains XbaI and NdeI sites for cloning purposes. PCR conditions were as described for the first reaction. The approximate 660 bp PCR product was digested with XbaI and BamHI, gel-purified and cloned into similarly cut plasmid pCDNA3.1 (+) (Invitrogen, Inc. Carlsbad, Calif.). Clone pCDNA3.1(+)::stII-GH(5C) or “5C” was determined to have the correct DNA sequence.
  • Clone “5C” was cleaved with NdeI and BarmHI and cloned into similarly cut pBBT108 (a derivative of pUC19 which lacks a Pst I site, this plasmid is described below). A clone with the correct insert was identified following digestion with these enzymes. This clone, designated pBBT111, was digested with NdeI and SalI, the 660 bp fragment containing the stII-GH fusion gene, was gel-purified and closed into the plasmid expression vector pCYB1 (New England BioLabs) that had been digested with the same enzymes and phosphatased.
  • pBBT111 was digested with NdeI and SalI, the 660 bp fragment containing the stII-GH fusion gene, was gel-purified and closed into the plasmid expression vector pCYB1 (New England BioLabs) that had been digested with the same enzymes and phosphatased.
  • a recombinant plasmid containing the stII-GH insertion was identified by restriction endonuclease digestions.
  • One such isolate was chosen for further studies and was designated pBBT114.
  • This plasmid was transformed into E. coli strains JM109 or W3110 (available from New England BioLabs and the American Type Culture Collection).
  • Wild type GH clone LB6 (pUC19: wild type GH) was used as the template to construct a GH clone containing the E. coli ompA signal sequence (Movva et al 1980). Because of its length, the ompA sequence was added in two sequential PCR reactions.
  • reverse primer BB10 (SEQ ID NO: 28) 5′CGCGGATCCGATTAGAATCCACAGCTCCCCTC3′.
  • PCR reactions were as described for amplifying wild type GH except that approximately 4 ng of plasmid LB6 was used as the template rather than single-stranded cDNA and the PCR conditions were 96° C. for 3 minutes, 30 cycles of (95° C. for 1 minute; 63° C. for 30 seconds; 72° C. for 1 minute) followed by 72° C. for 10 minutes.
  • the approximate 630 bp PCR product was gel-purified using the Qiaex II Gel Extraction Kit (Qiagen, Inc), diluted 50-fold in water and 2 microliters used as template for the second PCR reaction.
  • the second PCR reaction used reverse primer BB10 and forward PrimerBB6: 5′CCCCGTCGACACATATGAAGAAGACAGCTATCGCGATTGCAGTGGCACTGGCTGGTTTC3′. (SEQ ID NO: 32)
  • PCR conditions were as described for the first reaction.
  • the approximate 660 bp PCR product was gel-purified, digested with Sal I and Bam H1 and cloned into pUC19 (New England BioLabs) which was cut with Sal I and Bam H1 or pCDNA3.1(+) (Invitrogen) which had been cut by Xho I and Bam H1 ( Sal I and Xho I produce compatible single-stranded overhangs).
  • Xho I and Bam H1 Sal I and Xho I produce compatible single-stranded overhangs.
  • pBBT112 In order to generate a correct ompA-GH fusion gene segments of two sequenced clones which contained different errors separated by a convenient restriction site were recombined and cloned into the pUC19-derivative that lacks the Pst I site (see pBBT108 described below).
  • the resulting plasmid termed pBBT112, carries the ompA-GH fusion gene cloned as an Nde I-Bam H1 fragment into these same sites in pBBT108.
  • This plasmid is designated pBBT112 and is used in PCR-based, site-specific mutagenesis of GH as described below.
  • pUC19 plasmid DNA was digested with Pst I and subsequently treated at 75 deg. C. with PFU DNA Polymerase (Stratagene) using the vendor-supplied reaction buffer supplemented with 200 uM dNTPs. Under these conditions the polymerase will digest the 3′ single-stranded overhang created by Pst I digestion but will not digest into the double-stranded region.
  • the net result will be the deletion of the 4 single-stranded bases which comprise the middle four bases of the Pst I recognition site.
  • the resulting molecule has double-stranded, i.e., “blunt”, ends.
  • the linear monomer was gel-purified using the Qiaex II Gel Extraction Kit (Qiagen, Inc). This purified DNA was treated with T4 DNA Ligase (New England BioLabs) according to the vendor protocols, digested with Pst I, and used to transform E. coli DH5alpha. Transformants were picked and analyzed by restriction digestion with Pst I and Bam H1. One of the transformants which was not cleaved by Pst I but was cleaved at the nearby Bam H1 site was picked and designated pBBT108.
  • GH muteins were generally constructed using site-directed PCR-based mutagenesis as described in PCR Protocols: Current Methods and Applications edited by B. A. White, 1993 Humana Press, Inc., Totowa, N.J. and PCR Protocols: A Guide to Methods and Applications edited by Innis, M. A. et al 1990 Academic Press Inc San Diego, Calif.
  • PCR primer oligonucleotides are designed to incorporate nucleotide changes to the coding sequence of GH that result in substitution of a cysteine residue for an amino acid at a specific position within the protein.
  • Such mutagenic oligonucleotide primers can also be designed to incorporate an additional cysteine residue at the carboxy terminus or amino terminus of the coding sequence of GH. In this latter case one or more additional amino acid residues could also be incorporated at the amino terminal and/or carboxy terminal to the added cysteine residue if that were desirable.
  • oligonucleotides can be designed to incorporate cysteine residues as insertion mutations at specific positions within the GH coding sequence if that were desirable. Again, one or more additional amino acids could be inserted along with the cysteine residue and these amino acids could be positioned at the amino terminal and/or carboxy terminal to the cysteine residue.
  • the cysteine substitution mutation T135C was constructed as follows.
  • [0066] was designed to change the codon ACT for threonine at amino acid residue 135 to a TGT codon encoding cysteine and to span the nearby Bgl II site.
  • This oligonucleotide was used in PCR along with the forward oligonucleotide BB34 5′GTAGCGCAGGCCTTCCCAACCATT3′ (SEQ ID NO: 34) which anneals to the junction region of the ompA-GH fusion gene and is not mutagenic.
  • the PCR was performed in a 50 ul reaction in 1 ⁇ PCR buffer (Perkin-Elmer buffer containing 1.5 mM MgCl 2 ), 200 micromolar concentration of each of the four nucleotides dA, dC, dG and dT, with each oligonucleotide primer present at 0.5 ⁇ M, 5 pg of pBBT112 (described above) as template and 1.25 units of Amplitac DNA Polymerase (Perkin-Elmer) and 0.125 units of PFU DNA Polymerase (Stratagene). Reactions were performed in a Robocycler Gradient 96 thermal cycler (Stratagene). The program used entailed: 95 deg C.
  • the PCR reactions were analyzed by agarose gel electrophoresis to identify annealing temperatures that gave significant product of the expected size; ⁇ 430 bp.
  • the 45-deg C. reaction was “cleaned up” using the QIAquick PCR Purification Kit (Qiagen), digested with Bgl II and Pst I.
  • the resulting 278 bp BE II-Pst I fragment which includes the putative T135C mutation, was gel-purified and ligated into pBBT111 the pUC19 derivative carrying the stII-GH fusion gene (described above) which had been digested with Bgl II and Pst I and gel-purified.
  • Transformants from this ligation were initially screened by digestion with Bgl II and Pst I and subsequently one clone was sequenced to confirm the presence of the T135C mutation and the absence of any additional mutations that could potentially be introduced by the PCR reaction or by the synthetic oligonucleotides. The sequenced clone was found to have the correct sequence.
  • substitution mutation S132C was constructed using the protocol described above for T135C with the following differences: mutagenic reverse oligonucleotide BB29 5′CTGCTTGAAGATCTGCCCAGTCCGGGGGCAGCCATCTTC3′ (SEQ ID NO: 35) was used instead of BB28 and the PCR reaction with annealing temperature of 50 deg C. was used for cloning. One of two clones sequenced was found to have the correct sequence.
  • the substitution mutation T148C was constructed using an analogous protocol but employing a different cloning strategy.
  • the mutagenic forward oligonucleotide BB30 5′GGGCAGATCTTCAAGCAGACCTACAGCAAGTTCGACTGCAACTCACACAAC3′ (SEQ ID NO: 36) was used in PCR with the non-mutagenic reverse primer BB33 5′CGCGGTACCCGGGATCCGATTAGAATCCACAGCT3′ (SEQ ID NO: 37) which anneals to the most 3′ end of the GH coding sequence and spans the Bam H1 site immediately downstream.
  • PCR was performed as described above with the exception that the annealing temperatures used were 46, 51 and 56 deg C.
  • this cloning step is not orientation specific. Five of six clones tested were shown to be correctly oriented. One of these was sequenced and was shown to contain the desired T148C mutation. The sequence of the remainder of the 188 bp Bam H1-Bgl II mutagenic PCR fragment in this clone was confirmed as correct.
  • substitution mutation S144C was identical to the construction of T148C with the following exceptions.
  • Mutagenic forward oligonucleotide BB31 5′GGGCAGATCTTCAAGCAGACCTACTGCAAGTTCGAC3′ (SEQ ID NO: 38) was used instead of BB30.
  • Two of six clones tested were shown to be correctly oriented. One of these was sequenced and was shown to contain the desired S144C mutation.
  • the sequence of the remainder of the 188 bp Bam H1-Bgl II mutagenic PCR fragment in this clone was confirmed as correct.
  • a mutation was also constructed that added a cysteine residue to the natural carboxy terminus of GH.
  • the reverse mutagenic oligonucleotide BB32 5′CGCGGTACCGGATCCTTAGCAGAAGCCACAGCTGCCCTCCAC3′ (SEQ ID NO: 39) which inserts a TGC codon for cysteine between the codon for the carboxy terminal phe residue of GH and the TAA translational stop codon and spans the nearby Bam H1 site was used along with BB34 5′GTAGCGCAGGCCTTCCCAACCATT3′ (SEQ ID NO: 40) which is described above.
  • the 46 deg C. reaction was used for cloning. Three of six clones tested were shown to be correctly oriented. One of these was sequenced and was shown to contain the desired stp192C mutation. The sequence of the remainder of the 188 bp Bam H1-Bal II mutagenic PCR fragment in this clone was confirmed as correct.
  • Analogous PCR mutagenesis procedures can be used to generate other cysteine mutations.
  • the choice of sequences for mutagenic oligonucleotides will be dictated by the position where the desired cysteine residue is to be placed and the propinquity of useful restriction endonuclease sites.
  • Appropriate annealing temperatures for any oligonucleotide can be determined empirically.
  • the mutagenic oligonucleotide it is also desirable for the mutagenic oligonucleotide to span a unique restriction site so that the PCR product can be cleaved to generate a fragment that can be readily cloned into a suitable vector, e.g., one that can be used to express the mutein or provides convenient restriction sites for excising the mutated gene and readily cloning it into such an expression vector.
  • mutation sites and restriction sites are separated by distances that are greater than that which is desirable for synthesis of synthetic oligonucleotides: it is generally desirable to keep such oligonucleotides under 80 bases in length and lengths of 30-40 bases are more preferable.
  • genes targeted for mutagenesis could be re-engineered or re-synthesized to incorporate restriction sites at appropriate positions.
  • variations of PCR mutagenesis protocols employed above such as the so-called “Megaprimer Method” (Barik, S., pp. 277-286 in Methods in Molecular Biology, Vol. 15: PCR Protocols: Current Methods and Applications edited by B. A. White, 1993, Humana Press, Inc., Totowa, N.J.) or “Gene Splicing by Overlap Extension” (Horton, R. Mi., pp. 251-261, in Methods in Molecular Biology, Vol. 15: PCR Protocols: Current Methods and Applications, edited by B. A. White, 1993, Humana Press, Inc., Totowa, N.J.) can also be employed to construct such mutations.
  • pBBT120 GH gene with no leader sequence cloned into the tac expression vector pCYB1
  • pBBT114 GH gene with stII leader sequence cloned into the tac expression vector pCYB1
  • JM109 and W3 110 E. coli strains JM109 and W3 110.
  • the parental vector pCYB1 was also transformed into JM109 and W3110.
  • BOB119 JM109 (pCYB1)
  • BOB130 W3110 (pCYB1)
  • BOB129 JM109 (pBBT120)
  • BOB133 W3110 (pBBT120)
  • BOB121 JM109 (pBBT114)
  • BOB132 W3110 (pBBT114)
  • strains were grown overnight at 37° C. in Luria Broth (LB) (Sambrook, et al 1989) containing 100 ⁇ g/ml ampicillin. These saturated overnight cultures were diluted to ⁇ 0.03 OD at A 600 in LB containing 100 ⁇ g/ml ampicillin and incubated at 37° C. in shake flasks in rotary shaker, typically at 250-300 rpm. ODs were monitored and IPTG was added to a final concentration of 0.5 mM when culture ODs reached ⁇ 0.25 ⁇ 0.5, typically between 0.3 and 0.4. Cultures were sampled typically at 1, 3, 5 and ⁇ 16 h post-induction.
  • LB Luria Broth
  • sample buffer 50 mM Tris-HCl (pH 6.8), 2% sodium lauryl sulfate, 10% glycerol, 0.1% bromphenol blue
  • Samples were boiled for ⁇ 10 minutes or heated to 95° C. for ⁇ 10 minutes. Samples were cooled to room temperature before being loaded onto SDS polyacrylamide gels or were stored at ⁇ 20° C. if not run immediately.
  • This material could also be quantitatively bound to, and eluted from, a Q-Sepharose column using conditions very similar to those described for recombinant human GH by Becker and Hsiung, 1986 (FEBS Lett 204 pp145-150).
  • BB3 has the sequence: (SEQ ID NO: 26) 5′-CCCCGGATCCGCCACCATGGATCTCTGGCAGCTGCTGTT-3′.
  • BB4 has the sequence: (SEQ ID NO: 27) 5′CCCCGTCGACTCTAGAGCTATTAAATACGTAGCTCTTGGG-3′.
  • the template was single-stranded cDNA prepared from human liver (commercially available from CLONTECH Laboratories).
  • Primers BB3 and BB4 contain BamHI and SalI restriction sites, respectively, for cloning purposes.
  • the 100 ⁇ l PCR reactions contained 2.5 ng of the single-stranded cDNA and 20 picomoles of each primer in 1 ⁇ PCR buffer (Perkin-Elmer buffer containing MgCl 2 ), 200 micromolar concentration of each of the four nucleotides dA, dC, dG and dT, 2.5 units of Taq polymerase (Perkin-Elmer) and 2.5 units of Pfu polymerase (Stratagene, Inc).
  • the PCR reaction conditions were: 96° C.
  • thermocycler employed was the Amplitron II Thermal Cycler (Thermolyne).
  • the approximate 1.9 kb PCR product was digested with BamHI and SalI and ligated with similarly cut plasmid pUC19 ( New England BioLabs). However, none of the transformants obtained from this ligation reaction contained the 1.9 kb PCR fragment. Leung et al (Nature 1987 330 pp537-543) also failed to obtain full-length cDNA clones of the human GH receptor in pUC19.
  • PCR fragment was cloned into a low copy number vector, pACYC184 (New England BioLabs) at the BamHI and SalI sites in this vector.
  • pACYC184 New England BioLabs
  • Such clones were obtained at reasonable frequencies but E coli strains carrying the cloned PCR fragment grew poorly, forming tiny and heterogeneous looking colonies, in the presence of chloramphenicol, which is used to select for maintenance of pACYC 184.
  • PCR fragment was simultaneously cloned into pCDNA3.1 (+) (Invitrogen).
  • the approximate 1.9 kb PCR product was digested with BamHI and SalI and ligated into the BamHI and XhoI cloning sites of pCDNA3.1 (+).
  • Only infrequent transformants from this ligation contained the cloned GH receptor cDNA and all of those were found to contain deletions of segments of the receptor coding sequence.
  • One of these clones was sequenced and found to contain a deletion of 135 bp within the GH receptor coding sequence: the sequence of the rest of the gene was in agreement with that reported by Leung et at (1987).
  • the rabbit GH receptor was cloned by PCR using forward primer BB3 (described above) and reverse primer BB36.
  • BB36 has the sequence (SEQ ID NO: 41) 5′CCCCGTCGACTCTAGAGCCATTAGATACAAAGCTCTTGGG3′
  • Rabbit liver poly(A) + mRNA was purchased from CLONTECH, Inc. and used as the substrate in first strand synthesis of single-stranded cDNA to produce template for PCR amplification.
  • First strand synthesis of single-stranded cDNA was accomplished using a 1 st Strand cDNA Synthesis Kit for RT-PCR (AMV) kit from Boehringer Mannheim Corp (Indianapolis, Ind.) according to the vendor protocols.
  • Parallel first strand cDNA syntheses were performed using random hexamers or BB36 as the primer.
  • PCR reactions with the products of the first strand syntheses as templates, were carried out with primers BB3 and BB36 according to the 1 st Strand cDNA Synthesis Kit for RT-PCR (AMV) kit protocol and using 2.5 units of Amplitac DNA Polymerase (Perkin-Elmer) and 0.625 units of Pfu DNA Polymerase (Stratagene).
  • the PCR reaction conditions were 96° C. for 3 minutes, 35 cycles of (95° C., 1 minute; 58° C. for 30 seconds; 72° C. for 2 minutes), followed by 10 minutes at 72° C.
  • the thermocycler employed was the Amplitron II Thermal Cycler (Thermolyne).
  • the expected 1.9 kb PCR product was observed in PCR reactions using random hexamer-primed or BB36 primed cDNA as template.
  • the random hexamer-primed cDNA was used in subsequent cloning experiments. It was digested with Bam H1 and XbaI and run out over a 1.2% agarose gel. This digest generates two fragments ( ⁇ 365 bp and ⁇ 1600 bp) because the rabbit GH receptor gene contains an internal Bam H1 site. Both fragments were gel-purified. Initially the ⁇ 1600 bp Bam H1-XbaI fragment was cloned into pCDNA3.1(+) which had been digested with these same two enzymes.
  • one of the plasmids containing the 1600 bp Bam H1-Xba I fragment (pCDNA3.1(+):: rab-ghr-2A) was digested with Bam H1, treated with Calf Intestinal Alkaline Phosphatase (Promega) according to the vendor protocols, gel-purified and ligated with the gel purified ⁇ 365 bp Bam H1 fragment that contains the 5′ portion of the rabbit GH receptor gene.
  • the rabbit GH receptor can be employed in assays with human GH as a ligand as it has been shown that the human GH binds the rabbit receptor with high affinity (Leung et al 1987). Plasmids containing the cloned rabbit GH receptor should be sequenced to identify a rabbit GH receptor cDNA with the correct sequence before use.
  • a chimeric receptor could be constructed which combines the extracellular domain of the human receptor with the transmembrane and cytoplasmic domains of the rabbit receptor.
  • Such a chimeric receptor could be constructed by recombining the human and rabbit genes at the unique Nco I site that is present in each (Leung et al 1987).
  • Such a recombinant, containing the human gene segment located 5′ to, or “upstream” of the Nco I site and the rabbit gene segment 3′ to, or “downstream” of the Nco I site would encode a chimeric receptor of precisely the desired type, having the extracellular domain of the human receptor with the transmembrane and cytoplasmic domains of the rabbit receptor. This would allow analysis of the interaction of GH, GH muteins, and PEGylated GH muteins with the natural receptor binding site but could avoid the necessity of cloning the full length human GH receptor in E coli.
  • the GH muteins can be expressed in a variety of expression systems such as bacteria, yeast or insect cells.
  • Vectors for expressing GH muteins in these systems are available commercially from a number of suppliers such as Novagen, Inc. (pET15b for expression in E. coli ), New England Biolabs (pC4B1 for expression in E. coli ) Invitrogen (pVL1392, pVL1 393, and pMELBAC for expression in insect cells using Baculovirus vectors, Pichia vectors for expression in yeast cells, and pCDNA3 for expression in mammalian cells). Gn has been successfully produced in E.
  • coli as a cytoplasmic protein and as a secreted, periplasmic, protein using the E. coli OmpA or STII signal sequences to promote secretion of the protein into the periplasmic space (Chang et al., 1987; Hsiung et al., 1986). It is preferable that the GH muteins are expressed as secreted proteins so that they do not contain an N-terminal methionine residue, which is not present in the natural human protein.
  • DNA sequences encoding GH or GH muteins can be cloned into E. coli expression vectors such as pET15b that uses the strong T7 promoter or pCYB1 that uses the TAC promoter.
  • IPTG isopropylthiogalactopyranoside, available form Sigma Chemical Company
  • Adding IPTG can induce expression of the protein.
  • Recombinant GH will be secreted into the periplasmic space from which it can be released by, and subsequently purified, following osmotic shock (Becker and Hsiung, 1986).
  • the protein can be purified further using other chromatographic methods such as ion-exchange, hydrophobic interaction, size-exclusion and reversed phase chromatography, all of which are well known to those of skill in the art (e.g., see Becker and Hsiung, 1986). Protein concentrations can be determined using commercially available protein assay kits such as those sold by BioRad Laboratories (Richmond, Calif.). If the GH proteins are insoluble when expressed in E. coli they can be refolded using procedures well known to those skilled in the art (see Cox et al., 1994 and World patent applications WO9422466 and WO9412219).
  • the proteins can be expressed in insect cells as secreted proteins.
  • the expression plasmid can be modified to contain the GH signal sequence to promote secretion of the protein into the medium.
  • the cDNAs can be cloned into commercially available vectors, e.g., pVL1392 from Invitrogen, Inc., and used to infect insect cells.
  • the GH and GH muteins can be purified from conditioned media using conventional chromatography procedures.
  • Antibodies to rhGH can be used in conjunction with Western blots to localize fractions containing the GH proteins during chromatography.
  • fractions containing GH can be identified using ELISA assays.
  • the cysteine-added GH variants also can be expressed as intracellular or secreted proteins in eukaryotic cells such as yeast, insect cells or mammalian cells.
  • eukaryotic cells such as yeast, insect cells or mammalian cells.
  • Vectors for expressing the proteins and methods for performing such experiments are described in catalogues from various commercial supply companies such as Invitrogen, Inc., Stratagene, Inc. and ClonTech, Inc.
  • the GH and GH muteins can be purified using conventional chromatography procedures.
  • Biological activity of the GH muteins can be measured using a cell line that proliferates in response to GH.
  • Fuh et al. (1992) created a GH-responsive cell line by stably transforming a myeloid leukemia cell line, FDC-P1, with a chimeric receptor comprising the extracellular domain of the rabbit GH receptor fused to the mouse G-CSF receptor.
  • This cell line proliferates in response to GH with a half maximal effective concentration (EC 50 ) of 20 picomolar.
  • EC 50 half maximal effective concentration
  • a similar cell line can be constructed using the published sequences of these receptors and standard molecular biology techniques (Fuh et al., 1992).
  • the extracellular domain of the human GH receptor can be fused to the mouse G-CSF receptor using the published sequences of these receptors and standard molecular biology techniques.
  • Transformed cells expressing the chimeric receptor can be identified by flow cytometry using labeled GH, by the ability of transformed cells to bind radiolabeled GH, or by the ability of transformed cells to proliferate in response to added GH.
  • Purified GH and GH muteins can be tested in cell proliferation assays using cells expressing the chimeric receptor to measure specific activities of the proteins.
  • Cells can be plated in 96-well dishes with various concentrations of GH or GH muteins.
  • EC 50 can be determined for each mutein. Assays should be performed at least three times for each mutein using triplicate wells for each data point. GH muteins displaying similar optimal levels of stimulation and EC 50 values comparable to or greater than wild type GH are preferable.
  • GH muteins that retain in vitro activity can be PEGylated using a cysteine-reactive 8 kDa PEG-maleimide (or PEG-vinylsulfone) commercially available from Shearwater, Inc.
  • cysteine-reactive 8 kDa PEG-maleimide or PEG-vinylsulfone
  • methods for PEGylating the proteins with these reagents will be similar to those described in world patent applications WO 9412219 and WO 9422466 and PCT application US95/06540, with minor modifications.
  • the recombinant proteins must be partially reduced with dithiothreitol (DTT) in order to achieve optimal PEGylation of the free cysteine.
  • DTT dithiothreitol
  • the free cysteine is not involved in a disulfide bond, it is relatively unreactive to cysteine-reactive PEGs unless this partial reduction step is performed.
  • the amount of DTT required to partially reduce each mutein can be determined empirically, using a range of DTT concentrations. Typically, a 5-10 fold molar excess of DTT for 30 min at room temperature is sufficient. Partial reduction can be detected by a slight shift in the elution profile of the protein from a reversed-phase column. Care must be taken not to over-reduce the protein and expose additional cysteine residues.
  • Over-reduction can be detected by reversed phase-HPLC (the protein will have a retention time similar to the fully reduced and denatured protein) and by the appearance of GH molecules containing two PEGs (detectable by a molecular weight change on SDS-PAGE). Wild type GH can serve as a control since it should not PEGylate under similar conditions. Excess DTT can be removed by size exclusion chromatography using spin columns. The partially reduced protein can be reacted with various concentrations of PEG-maleimide (PEG: protein molar ratios of 1:1, 5:1,10:1 and 50:1) to determine the optimum ratio of the two reagents.
  • PEG-maleimide PEG: protein molar ratios of 1:1, 5:1,10:1 and 50:1
  • PEGylation of the protein can be monitored by a molecular weight shift using sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE). The lowest amount of PEG that gives significant quantities of mono-pegylated product without giving di-pegylated product will be considered optimum (80% conversion to mono-pegylated product is considered good).
  • mono-PEGylated protein can be purified from non-PEGylated protein and unreacted PEG by size-exclusion or ion exchange chromatography. The purified PEGylated protein can be tested in the cell proliferation assay described above to determine its specific activity.
  • Experiments can be performed to confirm that the PEG molecule is attached to the protein at the proper site. This can be accomplished by proteolytic digestion of the protein, purification of the PEG peptide (which will have a large molecular weight) by size exclusion, ion exchange or reversed phase chromatography, followed by amino acid sequencing or mass spectroscopy. The PEG-coupled amino acid will appear as a blank in the amino acid sequencing run.
  • the pharmacokinetic properties of the PEG-GH proteins can be determined as follows, or as described in world patent application WO9422466. Pairs of rats or mice can receive an intravenous bolus injection of the test proteins. Circulating levels of the proteins are measured over the course of 24 h by removing a small sample of blood from the animals at desired time points. Circulating levels of the test proteins can be quantitated using ELISA assays. Additional experiments can be performed using the subcutaneous route to administer the proteins. Similar experiments should be performed with the non-PEGylated protein to serve as a control. These experiments will reveal whether attachment of a PEG reagent to the protein alters its pharmacokinetic properties. Covalent modification of the protein with PEG should increase the protein's circulating half-life relative to the unPEGylated protein. Larger PEG molecules and/or attachment of multiple PEG molecules should lengthen the circulating half-life longer than smaller PEG molecules.
  • PEG-GH proteins can be tested in rodent models of growth hormone deficiency (Cox et al., 1994) and cachexia (Tomas et al., 1992; Read et al., 1992) to determine optimum dosing schedules and demonstrate efficacy. These studies can explore different size PEG molecules, e.g., 8 and 20 kDa, and dosing schedules to determine the optimum PEG size and dosing schedule. It is expected that the larger PEG molecule will increase the circulating half-life greater than the smaller PEG molecule and will require less frequent dosing.
  • GH deficiency model that can be used is a hypophysectomized rat.
  • GH stimulates body weight gain and bone and cartilage growth in this model (Cox et al., 1994).
  • Hypophysectomized rats can be purchased from Charles River. Rats can be injected with GH, PEG-GH or placebo and weight gain measured daily over a 10-14 day period. At time of sacrifice, tibial epiphysis width can be determined as a measure of bone growth. Experimental methods for performing these studies are described in Cox et al. (1994).
  • EPO erythropoietin
  • EPO is the hormone primarily responsible for stimulating erythropoiesis or red blood cell formation. EPO acts on immature red blood cell precursors to stimulate their further proliferation and differentiation into mature red blood cells.
  • a commercial pharmaceutical version is available from Amgen, Inc.
  • Human EPO is a 35-39 kDa glycoprotein secreted by the adult kidney. The mature human protein contains 166 amino acids and is heavily glycosylated.
  • SEQ ID NO: 2 The sequence of human EPO (SEQ ID NO: 2) is shown in Lin et al 1985 and Jacobs et al. 1985, which are incorporated herein by reference.
  • EPO The primary sequence of EPO is highly conserved among species (greater than 80% identity; Wen et al., 1994). Sugar groups account for greater than 40% of the protein's mass.
  • Human EPO contains three N-linked glycosylation sites and one O-linked glycosylation site. The N-linked glycosylation sites are conserved in different species whereas the O-linked glycosylation site is not. The extensive glycosylation of EPO has prevented the protein's crystallization, so the X-ray structure of the protein is not known.
  • Human EPO contains four cysteine residues. The disulfide assignments are Cys7 to Cys161 and Cys29 to Cys33.
  • Cys33 is not conserved in mouse EPO, suggesting that the Cys29-Cys33 disulfide bond is not critical to mouse EPO's structure or function. This conclusion also seems to hold for human EPO (Boissel et al., 1993).
  • EPO amino acid sequence of EPO is consistent with the protein being a member of the GH supergene family and mutational studies support this view of EPO's structure (Boissel et al., 1993; Wen et al., 1994). A model of the three dimensional structure of EPO, modeled after the GH structure has been proposed (Boissel et al., 1993; Wen et al., 1994).
  • Human EPO contains three sites for N-linked glycosylation (asparagine-24,-38 and -83) and one site for O-linked glycosylation (serine-126).
  • the N-linked glycosylation sites are located in the A-B and B-C loops and the O-glycosylation site is located in the C-D loop.
  • the N-linked glycosylation sites are conserved among species whereas the O-linked glycosylation site is adsent in rodent EPO (Wen et al., 1993).
  • a non-O-linked glycosylated human variant containing methionine at position 126 has been described (U.S. Pat. No. 4,703,008).
  • N-linked sugar groups are heavily branched and contain terminal sialic acid residues (Sasaki et al., 1987; Takeuchi et al., 1988).
  • N-38 and N-83 contain the most highly branched oligosaccharides (Sasaki et al., 1988).
  • the terminal sialic residues on EPO are critical for the protein's in vivo function because removal of these residues by digestion eliminates in vivo activity (Fukada et al., 1989; Spivak and Hogans, 1989). Loss of activity correlates with faster clearance of the asialated protein from the body.
  • the circulating half-life of the asialated protein in rats is less than ten minutes, in contrast to that of the sialated protein, which is approximately 2 hr (Fukada et al., 1989; Spivak and Hogans, 1989).
  • in vivo activity of EPO directly correlates with its circulating half-life.
  • N-linked sugars in EPO's biological activities haS been better defined by mutating, individually and in combination, the three asparagine residues comprising the N-linked glycosylation sites.
  • EPO muteins in which only a single N-linked glycosylation site was mutated, i.e., N24Q, N38Q and N83Q, were secreted from mammalian cells as efficiently as wild type EPO, indicating that N-linked glycosylation at all three sites is not required for protein secretion (N24Q indicates that the asparagine at position 24 is mutated to glutamine).
  • EPO muteins in which two or more N-linked glycosylation sites were mutated were secreted less efficiently than wild type EPO from mammalian cells (Yamaguchi et al., 1991; Delorme et al., 1992). Mutagenesis studies found that each of the single N-linked glycosylation site muteins had in vitro biological activities equal to or greater than wild type EPO. Thus, it was concluded that none of the N-linked glycosylation sites is essential for secretion or in vitro biological activity of EPO. In fact, removal of one of the glycosylation sites seemed to improve biological activity (Yamaguchi et al., 1991).
  • N-linked glycosylation muteins were studied by two groups. Yamaguchi et al. (1991) concluded that the N24Q and N83Q muteins had in vivo activities greater than wild type EPO, which correlated with their increased in vitro activities. These authors found that the N38Q mutein had decreased in vivo activity, about 60% of wild type EPO. N38 is the most heavily branched of the three N-linked glycosylation sites (Sasaki et al., 1988). Delorme et al. (1992) reported that mutating any of the N-linked glycosylation sites reduced in vivo biological activity by about 50%. Muteins in which two or more glycosylation sites were mutated had decreased in vivo activities in both studies.
  • N-linked glycosylation is required for in vitro and in vivo activity of EPO. Individually, however, none of the three glycosylation sites is absolutely essential for activity.
  • the N-linked sugars increase the apparent molecular weight of EPO and prolong its circulating half-life, which correlates with bioactivity.
  • Natural EPO and EPO manufactured in mammalian cells have complex N-linked sugars containing galactose and terminal sialic acid residues. The galactose residues are recognized by specific receptors on hepatocytes and promote rapid clearance of EPO from the body unless the galactose residues are masked by the terminal sialic acid residues.
  • This example provides for the design of cysteine-added EPO variants and their use in preparing conjugates using cysteine-reactive PEGs and other cysteine-reactive moieties.
  • Certain amino acids in EPO are non-essential for biological activity and can be mutated to cysteine residues without altering the normal disuifide binding patter, and overall conformation of the molecule.
  • amino acids are located in the A-B loop (amino acids 23-58 of the mature protein sequence), the B-C loop (amino acids 77-89 of the mature protein sequence), the C-D loop (amino acids 108-131 of the mature protein sequence), proximal to helix A (amino acids 1-8) and distal to helix D (amino acids 153-166 of the mature protein sequence). Also contemplated as preferred sites for adding cysteine residues are at the N-terminus or C-terminus of the protein sequence.
  • Preferred sites for cysteine substitutions are the O-linked glycosylation site (serine-126) and the amino acids comprising the three N-linked glycosylation sites (N24, I25, T26, N38, 139, T40, N83, S84, S85).
  • Glycosylation sites are attractive sites for introducing cysteine substitutions and attaching PEG molecules to EPO because (1) these sites are surface exposed; (2) the natural protein can tolerate bulky sugar groups at these positions; (3) the glycosylation sites are located in the putative loop regions and away from the receptor binding site (Wen et al., 1994); and (4) mutagenesis studies indicate these sites (at least individually) are not essential for in vitro or in vivo activity (Yamaguchi et al., 1991; Delorme et al., 1992). As discussed above, the local conformation of the region encompassing the O-glycosylation site region seems to be important for biological activity. Whether a cysteine substitution at position 126 affects biological activity has not been studied.
  • cysteine-29 to cysteine-33 disulfide bond is not necessary for biological activity of EPO because changing both residues to tyrosine simultaneously yielded a biologically active EPO protein (Boissel et al., 1993; Wen et al., 1994).
  • a “free” cysteine can be created by changing either cysteine-29 or cysteine-33 to another amino acid. Preferred amino acid changes would be to serine or alanine.
  • the remaining “free” cysteine (cysteine-29 or cysteine-33) would be a preferred site for covalently modifying the protein with cysteine-reactive moieties.
  • One aspect of the present invention is to provide expression systems in which the N24C, N38C and N83C EPO variants will have in vitro biological activities more similar to wild type EPO.
  • U.S. Pat. No. 4,703,008 contemplates naturally occurring variants of EPO as well as amino acid substitutions that are present in EPO proteins of mammals.
  • Ovine EPO contains a cysteine residue at position 88 of the polypeptide chain.
  • the inventor is unaware of any other naturally occurring human or animal cysteine variants of EPO in which the cysteine residue occurs in the polypeptide regions disclosed herein as being useful for generating cysteine-added EPO variants.
  • U.S. Pat. No. 4,703,008 specifically teaches away from cysteine-added EPO variants by suggesting that expression of EPO might be improved by deleting cysteine residues or substituting naturally occurring cysteine residues with serine or histidine residues.
  • the mature protein form of EPO can contain 165 or 166 amino acids because of post-translational removal of the C-terminal arginine. Asp-165 is the C-terminus of the 165 amino acid form and Agr-166 is the C-terminal amino acid of the 166 amino acid form.
  • the cysteine substitution and insertion mutations described herein can comprise either the 165 or 166 amino acid forms of mature EPO.
  • a cDNA encoding EPO can be cloned using the polymerase chain reaction (PCR) technique from the human HepG2 or Hep3B cell lines, which are known to express EPO when treated with hypoxia or cobalt chloride (Wen et al., 1993) and are available from the American Type Culture Collection (ATCC).
  • Cysteine mutations can be introduced into the cDNA by standard phage, plasmid or PCR mutagenesis procedures as described for GH.
  • the preferred sites for introduction of cysteine substitution mutations are in the A-B loop, the B-C loop, the C-D loop and the region proximal to helix A and distal to helix D.
  • the most preferred sites in these regions are the N— and O-linked glycosylation sites: S126C; N24C; I25C, T26C; N38C; I39C, T40C; N83C, S84C and S85C.
  • Other preferred sites for cysteine substitution mutagenesis are in the A-B loop, the B-C loop and the C-D loop, amino acids surrounding the glycosylation sites and the region of the protein proximal to helix A and distal to helix D (Boissel et al., 1993; Wen et al., 1994).
  • cysteine substitutions in these regions are: A1, P2, P3, R4, D8, S9, T27, G28, A30, E31, H32, S34, N36, D43, T44, K45, N47, A50, K52, E55, G57, Q58, G77, Q78, A79, Q86, W88, E89, T107, R110, A111, G113, A114, Q115, K116, E117, A118, S120, P121, P122, D123, A124, A125, A127, A128, T132, K154, T157, G158, E159, A160, T163, G164, D165, and R166.
  • Cysteine residues also can be introduced proximal to the first amino acid of the mature protein, i.e., proximal to A1, or distal to the final amino acid in the mature protein, i.e., distal to D165 or R166.
  • Wild type EPO and EPO muteins can be expressed using insect cells to determine whether the cysteine-added muteins are biologically active.
  • DNAs encoding EPO/EPO muteins can be cloned into the Baculovirus expression vector pVL1392 (available from Invitrogen, Inc. and Sigma Corporation (St. Louis, Mo.) and used to infect insect cells.
  • Recombinant Baculoviruses producing EPO can be identified by Western blots of infected insect cell conditioned media using polyclonal anti-human EPO antiserum (available from R&D Systems).
  • the secreted EPO mutein proteins can be purified by conventional chromatographic procedures well known to those of skill in the art. Protein concentrations can be determined using commercially available protein assay kits or ELISA assay kits (available from R&D Systems and Bio-Rad Laboratories).
  • EPO and EPO muteins can be tested in cell proliferation assays using EPO-responsive cell lines such as UT7-epo (Wen et al., 1994) or TF1 (available from the ATCC) to measure specific activities of the proteins.
  • Cells can be plated in 96-well microtiter plates with various concentrations of EPO.
  • Assays should be performed in triplicate. After 1-3 days in culture, cell proliferation can be measured by 3 H-thymidine incorporation, as described above for GH. The concentration of protein giving half-maximal stimulation (EC 50 ) can be determined for each mutein. Assays should be performed at least three times for each mutein, with triplicate wells for each data point.
  • EC 50 values can be used to compare the relative potencies of the muteins.
  • cell proliferation in response to added EPO muteins can be analyzed using an MTT dye-exclusion assay ( Komatsu et al. 1991). Proteins displaying similar optimal levels of stimulation and EC 50 values comparable to or greater than wild type EPO are preferable.
  • PEGylated EPO muteins that retain in vitro bioactivity, are candidates for testing in animal disease models. PEGylation of the protein at the proper amino acid can be determined as described for GH.
  • PEGylated EPO muteins expressed using insect cells may require that they be re-engineered for expression in mammalian cells to ensure proper glycosylation.
  • PEG-EPO candidates produced using insect cells can be tested in the animal models described below to determine if they are active in vivo and whether they are as active as PEG-EPO produced using mammalian cell expression systems.
  • the EPO muteins can be subdloned into commercially available eukaryotic expression vectors and used to stably transform Chinese Hamster Ovary (CHO) cells (available from the ATCC). Sublines can be screened for EPO expression using ELISA assays. Sufficient quantities of the insect cell- and mammalian cell-produced EPO muteins can be prepared to compare their biological activities in animal anemia models.
  • mice On day four or six after return to normal air pressure, mice are injected with erythropoietin or saline. Mice receive one injection per day for one to two days. One day later the animals receive an intravenous injection of labeled iron-59. The mice are euthanized 20 h later and the amount of labeled iron incorporated into red blood cells determined. EPO stimulates red blood cell formation in both models as measured by a dose-dependent increase in labeled iron incorporated into red blood cells. In both models different dosing regimens and different times of injections can be studied to determine if PEG-EPO is biologically active and/or more potent and produces longer acting effects than natural EPO.
  • Alpha interferon is produced by leukocytes and has antiviral, anti-tumor and immunomodulatory effects. There are at least 20 distinct alpha interferon genes that encode proteins that share 70% or greater amino acid identity. Amino acid sequences of the known alpha interferon species are given in Blatt et al. (1996). A “consensus” interferon that incorporates the most common amino acids into a single polypeptide chain has been described (Blatt et al., 1996). A hybrid alpha interferon protein may be produced by splicing different parts of alpha interferon proteins into a single protein (Horisberger and Di Marco, 1995).
  • alpha interferons contain N-linked glycosylation sites in the region proximal to helix A and near the B-C loop (Blatt et al. 1966).
  • the alpha 2 interferon protein (SEQ ID NO: 3) contains four cysteine residues that form two disulfide bonds.
  • the cys1-cys98 disulfide bond (cys1-cys99 in some alpha interferon species such as alpha 1; SEQ ID NO: 4) is not essential for activity.
  • the alpha 2-interferon protein does not contain any N-linked glycosylation sites.
  • the crystal structure of alpha interferon has been determined (Radhakrishnan et al., 1996).
  • This example provides cysteine added variants in the region proximal to the A helix, distal to the E helix, in the A-B loop, in the B-C loop, in the C-D loop and in the D-E loop.
  • Preferred sites for the introduction of cysteine residues in these regions of the alpha interferon-2 species are: D2, L3, P4, Q5, T6, S8, Q20, R22, K23, S25, F27, S28, K31, D32, R33, D35, G37, F38, Q40, E41, E42, F43, G44, N45, Q46, F47, Q48, K49, A50, N65, S68, T69, K70, D71, S72, S73, A74, A75, D77, E78, T79, Y89, Q90, Q91, N93, N94, E96, A97, Q101, G102, G104, T106, E107, T108, P109, K112, E113, D114, S115, K131, K132, K133, K134, Y135, S136, A139, S152, S154, T155, N156, L157, Q158, E159, S160, L161, R162, S163, K
  • cysteine residues are introduced proximal to the first amino acid of the mature protein, i.e., proximal to C1, or distal to the final amino acid in the mature protein, i.e., distal to E165 are provided.
  • Cys-1 has been deleted also are provided.
  • the cysteine variants may be in the context of any naturally occurring or non-natural alpha interferon sequence, e.g., consensus interferon or interferon protein hybrids.
  • alpha interferon species e.g., alpha interferon-1
  • alpha interferon-1 contain a naturally occurring “free” cysteine.
  • the naturally occurring free cysteine can be changed to another amino acid, preferably serine or alanine.
  • This example also provides cysteine variants of other alpha interferon species, including consensus interferon, at equivalent sites in these proteins.
  • the alignment of the alpha interferon-2 species with other known alpha interferon species and consensus interferon is given in Blatt et al. (1996).
  • the crystal structure of alpha interferon-2 has been determined by Rhadhakrishnan et al. (1996). Lydon et al (1985) found that deletion of the first four amino acids from the N-terminus of alpha interferon did not affect biological activity.
  • Valenzuela et al (1985) found that substitution of Phe-47 in alpha interferon-2 with Cys, Tyr or Ser did not alter biological activity of the protein. Cys-1 and Cys-98 have been changed individually to glycine and serine, respectively, without altering biological activity of the protein (DeChiara et al, 1986).
  • DNA sequences encoding alpha interferon-2 can be amplified from human genomic DNA, since alpha interferon genes do not contain introns (Pestka et al., 1987). The DNA sequence of alpha interferon-2 is given in Goeddel et al. (1980). Alternatively, a cDNA for alpha interferon-2 can be isolated from human lymphoblastoid cell lines that are known to express alpha interferon spontaneously or after exposure to viruses (Goeddell et al., 1980; Pickering et al., 1980). Many of these cell lines are available from the American Type Culture Collection (Rockville, Md.).
  • plasmid-based site-directed mutagenesis kits e.g., Quick-Change Mutagenesis Kit, Stratagene, Inc.
  • phage mutagenesis strategies or employing PCR mutagenesis as described for GH.
  • Alpha interferon has been successfully produced in E. coli as an intracellular protein (Tarnowski et al., 1986; Thatcher and Panayotatos, 1986). Similar procedures can be used to express alpha interferon muteins. Plasmids encoding alpha interferon or alpha interferon muteins can be cloned into an E. coli expression vector such as pET15b (Novagene, Inc.) that uses the strong T7 promoter or pCYB 1 (New England BioLabs, Beverly, Mass.) that uses the TAC promoter. Expression of the protein can be induced by adding IPTG to the growth media.
  • E. coli expression vector such as pET15b (Novagene, Inc.) that uses the strong T7 promoter or pCYB 1 (New England BioLabs, Beverly, Mass.) that uses the TAC promoter. Expression of the protein can be induced by adding IPTG to the growth media.
  • Recombinant alpha interferon expressed in E. coli is sometimes soluble and sometimes insoluble (Tarnowski et al., 1986; Thatcher and Panayotatos, 1986). Insolubility appears to be related to the degree of overexpression of the protein.
  • Insoluble alpha interferon proteins can be recovered as inclusion bodies and renatured to a fully active conformation following standard oxidative refolding protocols (Thatcher and Panayotatos, 1986; Cox et al., 1994).
  • the alpha interteron proteins can be purified further using other chromatographic methods such as ion-exchange, hydrophobic interaction, size-exclusion and reversed phase resins (Thatcher and Panayotaos, 1986). Protein concentrations can be determined using commercially available protein assay kits (Bio-Rad Laboratories).
  • alpha interferon muteins can be expressed in insect cells as secreted proteins as described for GH.
  • the proteins can be modified to contain the natural alpha interferon signal sequence (Goeddell et al., 1980) or the honeybee mellitin signal sequence (Invitrogen, Inc.) to promote secretion of the proteins.
  • Alpha interferon and alpha interferon muteins can be purified from conditioned media using conventional chromatography procedures.
  • Antibodies to alpha interferon can be used in conjunction with Western blots to localize fractions containing the alpha interferon proteins during chromatography. Alternatively, fractions containing alpha interferon proteins can be identified using ELISAs.
  • Bioactivities of alpha interferon and alpha interferon muteins can be measured using an in vitro viral plaque reduction assay (Ozes et al., 1992; Lewis, 1995). Human HeLa cells can be plated in 96-well plates and grown to near confluency at 37° C. The cells are then washed and treated for 24 hour with different concentrations of each alpha interferon preparation. Controls should include no alpha interferon and wild type alpha interferon (commercially available from Endogen, Inc., Woburn, Mass.).
  • VSV Vesicular stomatitis virus
  • EMCV encephalomyocarditis virus
  • EC 50 values (the amount of protein required to inhibit the cytopathic effect of the virus by 50%) can be used to compare the relative potencies of the proteins.
  • Wild type alpha interferon-2 protects cells from the cytopathic effects of VSV and EMCV and has a specific activity of approximately 2 ⁇ 10 8 units/mg in this assay (Ozes et al., 1992).
  • Alpha interferon muteins displaying EC 50 values comparable to wild type Alpha Interferon are preferable.
  • Alpha interferon muteins that retain activity can be PEGylated using procedures is similar to those described for GH. Wild type alpha interferon-2 can serve as a control since it should not PEGylate under similar conditions. The lowest amount of PEG that gives significant quantities of mono-pegylated product without giving di-pegylated product should be considered optimum.
  • Mono-PEGylated protein can be purified from non-PEGylated protein and unreacted PEG by size-exclusion or ion exchange chromatography. The purified PEGylated proteins can be tested in the viral plaque reduction bioassay described above to determine their bioactivities. PEGylated alpha interferon proteins with bioactivities comparable to wild type alpha interferon are preferable. Mapping the PEG attachment site and determination of pharmacokinetic data for the PEGylated protein can be performed as described for GH.
  • PEG-alpha interferon muteins can be tested using tumor xenograft models in nude mice and viral infection models (Balkwill, 1986; Fish et al., 1986). Since PEG-alpha interferon bioactivity may be species-specific, one should confirm activity of the PEGylated protein using appropriate animal cell lines in in vitro virus plaque reduction assays, similar to those described above. Next, one should explore the effects of different dosing regimens and different times of injections to determine if PEG-alpha interferon is more potent and produces longer lasting effects than non-PEGylated alpha interferon.
  • novel alpha interferon-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • Beta interferon is produced by fibroblasts and exhibits antiviral, antitumor and immunomodulatory effects.
  • the single-copy beta interferon gene encodes a preprotein that is cleaved to yield a mature protein of 166 amino acids (Taniguchi et al. 1980; SEQ ID NO: 5).
  • the protein contains three cysteines, one of which, cysteine-17, is “free”, i.e., it does not participate in a disulfide bond.
  • the protein contains one N-linked glycosylation site. The crystal structure of the protein has been determined (Karpusas et al. 1997)
  • This example provides cysteine-added variants at any of the three amino acids that comprise the N-linked glycosylation sites, i.e., N80C, E81C or T82C. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the E helix, in the A-B loop, in the B-C loop, in the C-D loop and in the D-E loop.
  • Preferred sites for introduction of cysteine residues in these regions are: M1, S2, Y3, N4, L5, Q23, N25, G26, R27, E29, Y30, K33, D34, R35, N37, D39, E42, E43, K45, Q46, L47, Q48, Q49, Q51, K52, E53, A68, F70, R71, Q72, D73, S74, S75, S76, T77, G78, E107, K108, E109, D110, F111, T112, R113, G114, K115, L116, A135, K136, E137, K138, S139, I157, N158, R159, L160, T161, G162, Y163, L164, R165 and N166.
  • cysteine residues are introduced proximal to the first amino acid of the mature protein, i.e., proximal to M1, or distal to the final amino acid in the mature protein, i.e., distal to N166 also are provided.
  • variants are produced in the context of the natural protein sequence or a variant protein in which the naturally occurring “free” cysteine residue (cysteine-17) has been changed to another amino acid, preferably serine or alanine.
  • novel beta interferon-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • G-CSF Granulocyte Colony-Stimulating Factor
  • G-CSF is a pleuripotent cytokine that stimulates the proliferation, differentiation and function of granulocytes.
  • the protein is produced by activated monocytes and macrophages.
  • the amino acid sequence of G-CSF (SEQ ID NO: 6) is given in Souza et al. (1986), Nagata et al. (1986a, b) and U.S. Pat. No. 4,810,643 all incorporated herein by reference.
  • the human protein is synthesized as a preprotein of 204 or 207 amino acids that is cleaved to yield mature proteins of 174 or 177 amino acids. The larger form has lower specific activity than the smaller form.
  • the protein contains five cysteines, four of which are involved in disulfide bonds.
  • Cysteine-17 is not involved in a disulfide bond. Substitution of cysteine-17 with serine yields a mutant G-CSF protein that is fully active (U.S. Pat. No. 4,810,643). The protein is O-glycosylated at threonine-133 of the mature protein.
  • A-B loop residues 40-70;
  • Helix B residues 71-91;
  • Helix C residues 100-123;
  • C-D loop residues 124-142
  • This example provides a cysteine-added variant at threonine-133.
  • This example provides other cysteine-added variants in the region proximal to helix A, distal to helix D, in the A-B loop, B-C loop and C-D loop.
  • Preferred sites for introduction of cysteine substitutions in these regions are: T1, P2, L3, G4, P5, A6, S7, S8, L9, P10, Q11, S12, T38, K40, S53, G55, W58, A59, P60, S62, S63, P65, S66, Q67, A68, Q70, A91, E93, G94, S96, E98, G100, G125, M126, A127, A129, Q131, T133, Q134, G135, A136, A139, A141, S142, A143, Q145, Q173 and P174.
  • cysteine residues are introduced proximal to the first amino acid of the mature protein, i.e., proximal to T1, or distal to the final amino acid in the mature protein, i.e., distal to P174 are provided.
  • These variants are provided in the context of the natural protein sequence or a variant protein in which the naturally occurring “free” cysteine residue (cysteine-17) has been changed to another amino acid, preferably serine or alanine.
  • a cDNA encoding human G-CSF can be purchased from R&D Systems (Minneapolis, Minn.) or amplified using PCR from mRNA isolated from human carcinoma cell lines such as 5637 and U87-MG known to express G-CSF constitutively (Park et al., 1989; Nagata, 1994). These cell lines are available from the American Type Culture collection (Rockville, Md.). Specific mutations can be introduced into the G-CSF sequence using plasmid-based site-directed mutagenesis kits (e.g., Quick-Change Mutagenesis Kit, Stratagene, Inc.), phage mutagenesis methods or employing PCR mutagenesis as described for GH.
  • plasmid-based site-directed mutagenesis kits e.g., Quick-Change Mutagenesis Kit, Stratagene, Inc.
  • phage mutagenesis methods employing PCR mutagenesis as described for GH.
  • G-CSF has been successfully produced in E. coli as an intracellular protein (Souza et al., 1986).
  • Plasmids encoding G-CSF or G-CSF muteins can be cloned into an E. coli expression vector such as pET15b (available from Novagen, Inc., Madison, Wis.) that uses the strong T7 promoter or pCYB1 (available from New England BioLabs, Beverly, Mass.) that uses the strong TAC promoter.
  • Expression of the protein can be induced by adding IPTG to the growth media.
  • Recombinant G-CSF expressed in E. coli is insoluble and can be recovered as inclusion bodies.
  • the protein can be renatured to a fully active conformation following standard oxidative refolding protocols (Souza et al., 1986; Lu et al., 1992; Cox et al., 1994). Similar procedures can be used to refold cysteine muteins.
  • the proteins can be purified further using other chromatographic methods such as ion exchange, hydrophobic interaction, size-exclusion and reversed phase resins (Souza et al., 1986; Kuga et al., 1989; Lu et al., 1992). Protein concentrations can be determined using commercially available protein assay kits (Bio-Rad Laboratories).
  • G-CSF and G-CSF muteins can be expressed in insect cells as secreted proteins as described for GH.
  • the proteins can be modified to contain the natural G-CSF signal sequence (Souza et al., 1986; Nagata et al., 1986a; Nagata et al, 1986b) or the honeybee mellitin signal sequence (Invitrogen, Inc., Carlsbad, Calif.) to promote secretion of the proteins.
  • G-CSF and G-CSF muteins can be purified from conditioned media using conventional chromatography procedures.
  • Antibodies to G-CSF can be used in conjunction with Western blots to localize fractions containing the G-CSF proteins during chromatography. Alternatively, fractions containing G-CSF proteins can be identified using ELISAs.
  • G-CSF muteins also can be expressed in mammalian cells as described for erythropoietin in Example 2.
  • Bioactivities of G-CSF and the G-CSF muteins can be measured using an in vitro cell proliferation assay.
  • the mouse NFS-60 cell line and the human AML-193 cell line can be used to measure G-CSF bioactivity (Tsuchiya et al., 1986; Lange et al., 1987; Shirafuji et al., 1989). Both cell lines proliferate in response to human G-CSF.
  • the AML-193 cell line is preferable since it is of human origin, which eliminates the possibility of a false conclusion resulting from species differences.
  • the NFS60 cell lines proliferates in response to G-CSF with a half-maximal effective concentration (EC 50 ) of 10-20 picomolar.
  • G-CSF and G-CSF muteins can be tested in cell proliferation assays using these cell lines to determinc specific activities of the proteins, using published methods (Tsuchiya et al., 1986; Lange et al., 1987; Shirafuji et al., 1989).
  • Cells can be plated in 96-well tissue culture dishes with different concentrations of G-CSF or G-CSF muteins. After 1-3 days at 37° C. in a humidified tissue culture incubator, proliferation can be measured by 3 H-thymidine incorporation as described for GH. Assays should be performed at least three times for each mutein using triplicate wells for each data point.
  • EC 50 values can be used to compare the relative potencies of the muteins.
  • G-CSF muteins displaying similar optimal levels of stimulation and EC 50 values comparable to wild type G-CSF are preferable.
  • G-CSF muteins that retain activity can be PEGylated using procedures similar to those described for GH. Wild type G-CSF and ser-17 G-CSF can serve as controls since they should not PEGylate under similar conditions. The lowest amount of PEG that gives significant quantities of mono-PEGylated product without giving di-PEGylated product should be considered optimum.
  • Mono-PEGylated protein can be purified from non-PEGylated protein and unreacted PEG by size-exclusion or ion exchange chromatography. The purified PEGylated proteins can be tested in the cell proliferation assay described above to determine their bioactivities.
  • the PEG site in the protein can be mapped using procedures similar to those described for GH.
  • Pharmacokinetic data for the PEGylated proteins can be obtained using procedures similar to those described for GH.
  • PEG-G-CSF The efficacy of PEG-G-CSF can be tested in a rat neutropenia model.
  • Neutropenia can be induced by treatment with cyclophosphamide, which is a commonly used chemotherapeutic agent that is myelosuppressive.
  • G-CSF accelerates recovery of normal neutrophil levels in cyclophosphamide-treated animals (Kubota et al., 1990). Rats receive an injection of cyclophosphamide on day 0 to induce neutropenia. The animals are then be divided into different groups, which will receive subcutaneous injections of G-CSF, PEG-G-CSF or placebo. Neutrophil and total white blood cell counts in peripheral blood should be measured daily until they return to normal levels.
  • novel GCSF-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • TPO Thrombopoietin
  • Thrombopoietin stimulates the development of megakaryocyte precursors of platelets.
  • the amino acid sequence of TPO (SEQ ID NO: 7) is given in Bartley et al.(1994), Foster et al.(l 994), de Sauvage et al.(1994), each incorporated herein by reference.
  • the protein is synthesized as a 353 amino acid precursor protein that is cleaved to yield a mature protein of 332 amino acids.
  • the N-terminal 154 amino acids have homology with EPO and other members of the GH supergene family.
  • the C-terminal 199 amino acids do not share homology with any other known proteins.
  • the C-terminal region contains six N-linked glycosylation sites and multiple O-linked glycosylation sites (Hoffman et al., 1996). O-linked glycosylation sites also are found in the region proximal to the A helix, in the A-B loop, and at the C-terminus of Helix C (Hoffman et al., 1996).
  • a truncated TPO protein containing only residues 1-195 of the mature protein is fully active in vitro (Bartley et al., 1994).
  • This example provides cysteine-added variants at any of the amino acids that comprise the N-linked glycosylation sites and the O-linked glycosylation sites. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop.
  • Preferred sites for introduction of cysteine residues are: S1, P2, A3, P4, P5, A6, T37, A43, D45, S47, G49, E50, K52, T53, Q54, E56, E57, T58, A76, A77, R78, G79, Q80, G82, T84, S87, S88, G109, T110, Q111, P113, P114, Q115, G116, R117, T118, T119, A120, H121, K122, G146, G147, S148, T149, A155, T158,T159, A160, S163, T165, S166, T170, N176, R177, T178, S179, G180, E183, T184, N185, F186, T187, A188, S189, A190, T192, T193, G194, S195, N213, Q214, T215, S216, S218, N234, G235, T236, S244, T247, S254, S
  • cysteine residues are introduced proximal to the first amino acid of the mature protein, i.e., proximal to S1, or distal to the final amino acid in the mature protein, i.e., distal to G332 are provided.
  • the cysteine-added variants are provided in the context of the natural human protein or a variant protein that is truncated between amino acids 147 and the C-terminus of the natural protein, G332.
  • cysteine residues are added distal to the final amino acid of a TPO protein that is truncated between amino acids 147 and 332 also are provided.
  • novel TPO-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • GM-CSF Granulocyte-Macrophage Colony Stimulating Factor
  • GM-CSF stimulates the proliferation and differentiation of various hematopoietic cells, including neutrophil, monocyte, eosinophil, erythroid, and megakaryocyte cell lineages.
  • the amino acid sequence of human GM-CSF (SEQ ID NO: 8) is given in Cantrell et al. (1985) and Lee et al (1985) both incorporated herein by reference.
  • GM-CSF is produced as a 144 amino acid preprotein that is cleaved to yield a mature 127 amino acid protein.
  • the mature protein has two sites for N-linked glycosylation. One site is located at the C-terminal end of Helix A; the second site is in the A-B loop.
  • This example provides cysteine-added variants at any of the amino acids that comprise the N-linked glycosylation sites, i.e., N27C, L28C, S29C, N37C, E38C and T39C. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop.
  • Preferred sites for introduction of cysteine substitutions in these regions are: A1, P2, A3, R4, S5, P6, S7, P8, S9, T10, Q11, R30, D31, T32, A33, A34, E35, E41, S44, E45, D48, Q50, E51, T53, Q64, G65, R67, G68, S69, L70, T71, K72, K74, G75, T91, E93, T94, S95, A97, T98, T102, I117, D120, E123, V125, Q126 and E127.
  • cysteine residues are introduced proximal to the first amino acid of the mature protein, i.e., proximal to A1, or distal to the final amino acid in the mature protein, i.e., distal to E127 are provided.
  • novel GM-CSF-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • IL-2 is a T cell growth factor that is synthesized by activated T cells.
  • the protein stimulates clonal expansion of activated T cells.
  • Human IL-2 is synthesized as a 153 amino acid precursor that is cleaved to yield a 133 amino acid mature protein (Tadatsugu et al., 1983; Devos et al., 1983; SEQ ID NO: 9).
  • the amino acid sequence of IL-2 is set forth in (Tadatsugu et al. 1983; Devos et al. 1983).
  • the mature protein contains three cysteine residues, two of which form a disulfide bond. Cysteine-125 of the mature protein is not involved in a disulfide bond. Replacement of cysteine-125 with serine yields an IL-2 mutein with full biological activity (Wang et al., 1984).
  • the protein is O-glycosylated at threonine-3 of the mature protein chain.
  • This example provides cysteine-added variants in the last four positions of the D helix, in the region distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop.
  • These variants are provided in the context of the natural protein sequence or a variant protein in which the naturally occurring “free” cysteine residue (cysteine-125) has been changed to another amino acid, preferably serine or alanine.
  • variants in which cysteine residues are introduced proximal to the first amino acid, i.e., A1, or distal to the final amino acid, i.e., T133, of the mature protein, also are provided.
  • novel IL-2-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • IL-3 is produced by activated T cells and stimulates the proliferation and differentiation of pleuripotent hematopoietic stem cells.
  • the amino acid sequence of human IL-3 (SEQ ID NO: 10) is given in Yang et al. (1986); Dorssers et al. (1987) and Otsuka et al. (1988) all incorporated herein by reference.
  • the protein contains two cysteine residues and two N-linked glycosylation sites. Two alleles have been described, resulting in isoforms having serine or proline at amino acid position 8 or the mature protein.
  • This example provides cysteine-added variants at any of the amino acids that comprise the N-linked glycosylation sites.
  • This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop.
  • Variants in which cysteine residues are introduced proximal to the first amino acid or distal to the final amino acid in the mature protein also are provided.
  • novel IL-3-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • IL-4 is a pleiotropic cytokine that stimulates the proliferation and differentiation of monocytes and T and B cells. IL-4 has been implicated in the process that leads to B cells secreting IgE, which is believed to play a role in asthma and atopy.
  • the bioactivity of IL-4 is species specific. IL-4 is synthesized as a 153 amino acid precursor protein that is cleaved to yield a mature protein of 129 amino acids.
  • the amino acid sequence of human IL-4 (SEQ ID NO: 11) is given in Yokota et al. (1986) which is incorporated herein by reference.
  • the protein contains six cysteine residues and two N-linked glycosylation sites. The glycosylation sites are located in the A-B and C-D loops.
  • This example provides cysteine-added variants at any of the amino acids that comprise the N-linked glycosylation sites.
  • This, example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop.
  • Variants in which cysteine residues are introduced proximal to the first amino acid, H1, or distal to the final amino acid, S129, of the mature protein are provided.
  • novel IL-4-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • IL-5 is a differentiation and activation factor for eosinophils.
  • the amino acid sequence of human IL-5 (SEQ ID NO: 12) is given in Yokota et al. (1987) which is incorporated herein by reference.
  • the mature protein contains 115 amino acids and exists in solution as a disulfide-linked homodimer.
  • the protein contains both O-linked and N-linked glycosylation sites.
  • This example provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid in the mature protein are provided.
  • novel IL-5-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • IL-6 stimulates the proliferation and differentiation of many cell types.
  • the amino acid sequence of human IL-6 (SEQ ID NO: 13) is given in Hirano et al. (1986) which is incorporated herein by reference.
  • IL-6 is synthesized as a 212 amino acid preprotein that is cleaved to generate a 184 amino acid mature protein.
  • the mature protein contains two sites for N-linked glycosylation and one site for O-glycosylation, at T137, T138, T142 or T143.
  • This example provides cysteine-added variants at any of the amino acids that comprise the N-linked glycosylation sites and at the O-linked glycosylation site. Also provided are cysteine-added variants in the region proximal to the A helix, distal to the D nelix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
  • novel IL-6-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • IL-7 stimulates proliferation of immature B cells and acts on mature T cells.
  • the amino acid sequence of human IL-7 (SEQ ID NO: 14) is given in Goodwin et al. (1989) which is incorporated herein by reference.
  • the protein is synthesized as a 177 amino acid preprotein that is cleaved to yield a 152 amino acid mature protein that contains three sites for N-linked glycosylation.
  • This example provides cysteine-added variants at any of the amino acids that comprise the N-linked glycosylation sites. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop.
  • novel IL-7-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
  • IL-9 is a pleiotropic cytokine that acts on many cell types in the lymphoid, myeloid and mast cell lineages. IL-9 stimulates the proliferation of activated T cells and cytotoxic T lymphocytes, stimulates proliferation of mast cell precursors and synergizes with erythropoietin in stimulating immature red blood cell precursors.
  • the amino acid sequence of human IL-9 (SEQ ID NO: 15) is given in Yang et al. (1989) which is incorporated herein by reference.
  • IL-9 is synthesized as a precursor protein of 144 amino acids that is cleaved to yield a mature protein of 126 amino acids. The protein contains four potential N-linked glycosylation sites.
  • This example provides cysteine-added variants at any of the three amino acids that comprise the N-linked glycosylation sites. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
  • novel IL-9-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • IL-10 The amino acid sequence of human IL-10 (SEQ ID NO: 16) is given in Vieira et al. (1991) which is incorporated herein by reference.
  • IL-10 is synthesized as a 178 amino acid precursor protein that is cleaved to yield a mature protein of 160 amino acids.
  • IL-10 can function to activate or suppress the immune system.
  • the protein shares structural homology with the interferons, i.e., it contains five amphipathic helices.
  • the protein contains one N-linked glycosylation site.
  • This example provides cysteine-added variants at any of the three amino acids comprising the N-linked glycosylation site.
  • This example also provides cysteine-added variants in the region proximal to the A helix, distal to the E helix, in the A-B loop, in the B-C loop, in the C-D loop and in the D-E loop.
  • Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
  • novel IL-10-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • IL-11 is a pleiotropic cytokine that stimulates hematopoiesis, lymphopoeisis and acute phase responses. IL-11 shares many biological effects with IL-6.
  • the amino acid sequence of human IL-11 (SEQ ID NO: 17) is given in Kawashima et al. (1991) and Paul et al. (1990) both incorporated herein by reference.
  • IL-11 is synthesized as a precursor protein of 199 amino acids that is cleaved to yield a mature protein of 178 amino acids. There are no N-linked glycosylation sites in the protein.
  • This example provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
  • novel IL-11-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • IL-12 stimulates proliferation and differentiation of NK cells and cytotoxic T lymphocytes.
  • IL-12 exists as a heterodimer of a p35 subunit and a p40 subunit.
  • the p35 subunit is a member of the GH supergene family.
  • the amino acid sequence of the p35 subunit (SEQ ID NO: 18) is given in Gubler et al. (1991) and Wolf et al. (1991) both incorporated herein by reference.
  • p35 is synthesized as a precursor protein of 197 amino acids and is cleaved to yield a mature protein of 175 amino acids.
  • the protein contains 7 cysteine residues and three potential N-linked glycosylation sites.
  • This example provides cysteine-added variants at any of the three amino acids that comprise the three N-linked glycosylation sites.
  • This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop.
  • These variants are provided in the context of the natural protein sequence or a variant protein in which the naturally occurring “free” cysteine residue has been changed to another amino acid, preferably serine or alanine.
  • Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
  • novel IL-12 p35-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • IL-13 shares many biological properties with IL-4.
  • the amino acid sequence of human IL-13 (SEQ ID NO: 19) is given in McKenzie et al. (1993) and Minty et al. (1993) both incorporated herein by reference.
  • the protein is synthesized as a 132 amino acid precursor protein that is cleaved to yield a mature protein of 112 amino acids.
  • the mature protein contains 5 cysteine residues and multiple N-linked glycosylation sites.
  • a variant in which glutamine at position 78 is deleted due to alternative mRNA splicing has been described (McKenzie et al 1993)
  • This example provides cysteine-added variants at any of the three amino acids comprising the N-linked glycosylation sites.
  • This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop.
  • These variants are provided in the context of the natural protein sequence or a variant sequence in which the pre-existing “free” cysteine has been changed to another amino acid, preferably to alanine or serine.
  • Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
  • novel IL-13-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • IL-15 stimulates the proliferation and differentiation of T cells, NK cells, LAK cells and Tumor Infiltrating Lymphocytes. IL-15 may be useful for treating cancer and viral infections.
  • the sequence of IL-15 (SEQ ID NO: 20) is given in Anderson et al. (1995) which is incorporated herein by reference.
  • IL-15 contains two N-linked glycosylation sites, which are located in the C-D loop and C-terminal end of the D helix.
  • IL-15 encodes a 162 amino acid preprotein that is cleaved to generate a mature 114 amino acid protein.
  • This example provides cysteine-added variants at any of the three amino acids comprising the N-linked glycosylation sites in the C-D loop or C-terminal end of the D helix.
  • This example also provides cysteine-added variants proximal to helix A, in the A-B loop, the B-C loop, the C-D loop or distal to helix D.
  • Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
  • novel IL-15-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • M-CSF Macrophage Colony Stimulating Factor
  • M-CSF regulates the growth, differentiation and function of monocytes.
  • the protein is a disulfide-linked homodimer. Multiple molecular weight species of M-CSF, which arise from differential mRNA splicing, have been described.
  • the amino acid sequence of human M-CSF and its various processed forms are given in Kawasaki et al (1985), Wong et al. (1987) and Cerretti et al (1988) which are incorporated herein by reference. Cysteine-added variants can be produced following the general teachings of this application and in accordance with the examples set forth herein.
  • novel MCSF-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • Oncostatin M is a multifunctional cytokine that affects the growth and differentiation of many cell types.
  • the amino acid sequence of human oncostatin M (SEQ ID NO: 21) is given in Malik et al. (1989) which is incorporated herein by reference.
  • Oncostatin M is produced by activated monocytes and T lymphocytes.
  • Oncostatin M is synthesized as a 252 amino acid preprotein that is cleaved sequentially to yield a 227 amino acid protein and then a 196 amino acid protein (Linsley et al., 1990).
  • the mature protein contains O-linked glycosylation sites and two N-linked glycosylation sites.
  • the protein is O-glycosylated at T160, T162 and S165.
  • the mature protein contains five cysteine residues.
  • This example provides cysteine-added variants at either of the three amino acids comprising the N-linked glycosylation sites or at the amino acids that comprise the O-linked glycosylation sites.
  • This example also provides cysteine-added variants proximal to helix A, in the A-B loop, in the B-C loop, in the C-D loop or distal to helix D.
  • These variants are provided in the context of the natural protein sequence or a variant sequence in which the pre-existing “free” cysteine has been changed to another amino acid, preferably to alanine or serine.
  • Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
  • CNTF Ciliary Neurotrophic Factor
  • CNTF is a 200 amino acid protein that contains no glycosylation sites or signal sequence for secretion.
  • the protein contains one cysteine residue.
  • CNTF functions as a survival factor for nerve cells.
  • This example provides cysteine-added variants proximal to helix A, in the A-B loop, the B-C loop, the C-D loop or distal to helix D.
  • These variants are provided in the context of the natural protein sequence or a variant sequence in which the pre-existing “free” cysteine has been changed to another amino acid, preferably to alanine or serine.
  • Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
  • novel CNTF-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • LIF Leukemia Inhibitory Factor
  • LIF LIF
  • the human gene encodes a 202 amino acid precursor that is cleaved to yield a mature protein of 180 amino acids.
  • the protein contains six cysteine residues, all of which participate in disulfide bonds.
  • the protein contains multiple O— and N-linked glycosylation sites.
  • the crystal structure of the protein was determined by Robinson et al. (1994). The protein affects the growth and differentiation of many cell types.
  • This example provides cysteine-added variants at any of the three amino acids comprising the N-linked glycosylation sites or the O-linked glycosylation site. Also provided are cysteine-added variants proximal to helix A, in the A-B loop, the B-C loop, the C-D loop or distal to helix D. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
  • novel LIF-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
  • the protein analogues disclosed herein can be used for the known therapeutic uses of the native proteins in essentially the same forms and doses all well known in the art.

Abstract

The growth hormone supergene family comprises greater than 20 structurally related cytokines and growth factors. A general method is provided for creating site-specific, biologically active conjugates of these proteins. The method involves adding cysteine residues to non-essential regions of the proteins or substituting cysteine residues for non-essential amino acids in the proteins using site-directed mutagenesis and then covalently coupling a cysteine-reactive polymer or other type of cysteine-reactive moiety to the proteins via the added cysteine residue. Disclosed herein are preferred sites for adding cysteine residues or introducing cysteine substitutions into the proteins, and the proteins and protein derivatives produced thereby.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 10/400,377, filed Mar. 26, 2003, which is a divisional of U.S. application Ser. No. 09/462,941, filed Jan. 14, 2000, now U.S. Pat. No. 6,608,183, which is a national stage application under 35 U.S.C. § 371 of PCT Application Serial No. PCT/US98/14497, filed Jul. 13, 1998, which claims the benefit of priority from U.S. Provisional Application Ser. No. 60/052,516, filed Jul. 14, 1997, each of which is incorporated herein by reference in its entirety.[0001]
  • GOVERNMENT SUPPORT
  • [0002] This invention was made in part with government support under Grant Nos. 1R43 CA78094-01 and 2R44 CA78094-02, each awarded by the National Institutes of Health. The government has certain rights in the invention.
  • FIELD OF THE INVENTION
  • The present invention relates to genetically engineered therapeutic proteins. More specifically, the engineered proteins include growth hormone and related proteins. [0003]
  • BACKGROUND OF THE INVENTION
  • The following proteins are encoded by genes of the growth hormone (GH) supergene family (Bazan (1990); Mott and Campbell (1995); Silvennoinen and lhle (1996)): growth hormone, prolactin, placental lactogen, erythropoietin (EPO), thrombopoietin (TPO), interleukin-2 (IL-2), IL-3, IL-4, IL-5, IL-6, IL-7, IL-9, IL-10, IL-11, IL-12 (p35 subunit) IL-13, IL-15, oncostatin M, ciliary neurotrophic factor, leukemia inhibitory factor, alpha interferon, beta interferon, gamma interferon, omega interferon, tau interferon, granulocyte-colony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating factor (M-CSF) and cardiotrophin-1 (CT-1) (“the GH supergene family”). It is anticipated that additional members of this gene family will be identified in the future through gene cloning and sequencing. Members of the GH supergene family have similar secondary and tertiary structures, despite the fact that they generally have limited amino acid or DNA sequence identity. The shared structural features allow new members of the gene family to be readily identified. [0004]
  • There is considerable interest on the part of patients and healthcare providers in the development of long acting, “user-friendly” protein therapeutics. Proteins are expensive to manufacture and, unlike conventional small molecule drugs, are not readily absorbed by the body. Moreover, they are digested if taken orally. Therefore, natural proteins must be administered by injection. After injection, most proteins are cleared rapidly from the body, necessitating frequent, often daily, injections. Patients dislike injections, which leads to reduced compliance and reduced drug efficacy. Some proteins, such as erythropoietin (EPO), are effective when administered less often (three times per week for EPO) because they are glycosylated. However, glycosylated proteins are produced using expensive mammalian cell expression systems. [0005]
  • The length of time an injected protein remains in the body is finite and is determined by, e.g., the protein's size and whether or not the protein contains covalent modifications such as glycosylation. Circulating concentrations of injected proteins change constantly, often by several orders of magnitude, over a 24-hour period. Rapidly changing concentrations of protein agonists can have dramatic downstream consequences, at times under-stimulating and at other times over-stimulating target cells. Similar problems plague protein antagonists. These fluctuations can lead to decreased efficacy and increased frequency of adverse side effects for protein therapeutics. The rapid clearance of recombinant proteins from the body significantly increases the amount of protein required per patient and dramatically increases the cost of treatment. The cost of human protein pharmaceuticals is expected to increase dramatically in the years ahead as new and existing drugs are approved for more disease indications. [0006]
  • Thus, there is a need to develop protein delivery technologies that lower the costs of protein therapeutics to patients and healthcare providers. The present invention provides a solution to this problem by providing methods to prolong the circulating half-lives of protein therapeutics in the body so that the proteins do not have to be injected frequently. This solution also satisfies the needs and desires of patients for protein therapeutics that are “user-friendly”, i.e., protein therapeutics that do not require frequent injections. The present invention solves these and other problems by providing biologically active, cysteine-added variants of members of the growth hormone supergene family. The invention also provides for the chemical modification of these variants with cysteine-reactive polymers or other types of cysteine-reactive moieties to produce derivatives thereof and the molecules so produced. [0007]
  • SUMMARY OF THE INVENTION
  • The present invention provides cysteine variants of members of the GH supergene family. The variants comprise a cysteine residue substituted for a nonessential amino acid of the proteins. Preferably, the variants comprise a cysteine residue substituted for an amino acid selected from amino acids in the loop regions, the ends of the alpha helices, proximal to the first amphipathic helix, and distal to the final amphipathic helix or wherein the cysteine residue is added at the N-terminus or C-terminus of the proteins. Preferred sites for substitution are the N— and O-linked glycosylation sites. [0008]
  • Also provided are cysteine variants wherein the amino acid substituted for is in the A-B loop, B-C loop, the C-D loop or D-E loop of interferon/interferon-10-like members of the GH supergene family. [0009]
  • Also provided are cysteine variants of members of the GH supergene family wherein the cysteine residue is introduced between two amino acids in the natural protein. In particular, the cysteine residue is introduced into the loop regions, the ends of the alpha helices, proximal to the first amphipathic helix, or distal to the final amphipathic helix. Even more particularly, the cysteine variant is introduced between two amino acids in an N—O-linked glycosylation site or adjacent to an amino acid in an N-linked or O-linked glycosylation site. [0010]
  • More particularly are provided cysteine variants wherein the loop region where the cysteine is introduced is the A-B loop, the B-C loop, the C-D loop or D-E loop of interferon/interferon-10-like members of the GH supergene family. [0011]
  • Such cysteine substitutions or insertion mutations also can include the insertion of one or more additional amino acids amino acids at the amino-terminal or carboxy-terminal to the cysteine substitution or insertion. [0012]
  • Also provided are cysteine variants that are further derivatised by PEGylating the cysteine variants and including the derivatised proteins produced thereby. [0013]
  • As set forth in the examples, specific cysteine variants of the members of the GH supergene family also are provided, including for example, variants of GH. The GH cysteine variants can have the substituted-for amino acid or inserted cysteine located at the N-terminal end of the A-B loop, the B-C loop, the C-D loop, the first three or last three amino acids in the A, B, C and D helices and the amino acids proximal to helix A and distal to helix D. [0014]
  • More particularly, the cysteine can be substituted for the following amino acids: F1, T3, P5, E33, A34, K38, E39, Q40, S43, Q46, N47, P48, Q49, T50, S51, S55, T60, A98, N99, S100, G104, A105, S106, E129, D130, G131, S132, P133, T135, G136, Q137, K140, Q141, T142, S144, K145, D147, T148, N149, S150, H151, N152, D153, S184, E186, G187, S188, and G190. [0015]
  • Other examples of cysteine variants according to the invention include erythropoietin variants. Erythropoietin variants include those wherein the substituted for amino acid is located in the A-B loop, the B-C loop, the C-D loop, the amino acids proximal to helix A and distal to helix D and the N— or C-terminus. Even more specifically, the EPO cysteine variants include molecules wherein the amino acids indicated below have a cysteine substituted therefor: serine-126, N24, I25, T26, N38, I39, T40, N83, S84, A1, P2, P3, R4, D8, S9, T27, G28, A30, E31, H32, S34, N36, D43, T44, K45, N47, A50, K52, E55, G57, Q58, G77, Q78, A79, Q86, W88, E89, T107, R110, A111, G113, A114, Q15, K116, E117, A118, S120, P121, P122, D123, A124, A125, A127, A128, T132, K154, T157, G158, E159, A160, T163, G164, D165, R166 and S85. [0016]
  • The members of the GH supergene family include growth hormone, prolactin, placental lactogen, erythropoietin, thrombopoietin, interleukin-2, interleukin-3, interleukin-4, interleukin-5, interleukin-6, interleukin-7, interleukin-9, interleukin-10, interleukin-1, interleukin-12 (p35 subunit), interleukin-13, interleukin-15, oncostatin M, ciliary neurotrophic factor, leukemia inhibitory factor, alpha interferon, beta interferon, gamma interferon, omega interferon, tau interferon, granulocyte-colony stimulating factor, granulocyte-macrophage colony stimulating factor, macrophage colony stimulating factor, cardiotrophin-1 and other proteins identified and classified as members of the family. The proteins can be derived from any animal species including human, companion animals and farm animals. [0017]
  • Other variations and modifications to the invention will be obvious to those skilled in the art based on the specification and the “rules” set forth herein. All of these are considered as part of the invention.[0018]
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to cysteine variants and, among other things, the site-specific conjugation of such proteins with polyethylene glycol (PEG) or other such moieties. PEG is a non-antigenic, inert polymer that significantly prolongs the length of time a protein circulates in the body. This allows the protein to be effective for a longer period of time. Covalent modification of proteins with PEG has proven to be a useful method to extend the circulating half-lives of proteins in the body (Abuchowski et al., 1984; Hershfield, 1987; Meyers et al., 1991). Covalent attachment of PEG to a protein increases the protein's effective size and reduces its rate of clearance rate from the body. PEGs are commercially available in several sizes, allowing the circulating half-lives of PEG-modified proteins to be tailored for individual indications through use of different size PEGs. Other benefits of PEG modification include an increase in protein solubility, an increase in in vivo protein stability and a decrease in protein immunogenicity (Katre et al., 1987; Katre, 1990). [0019]
  • The preferred method for PEGylating proteins is to covalently attach PEG to cysteine residues using cysteine-reactive PEGs. A number of highly specific, cysteine-reactive PEGs with different reactive groups (e.g., maleimide, vinylsulfone) and different size PEGs (2-20 kDa) are commercially available (e.g., from Shearwater, Polymers, Inc., Huntsville, Ala.). At neutral pH, these PEG reagents selectively attach to “free” cysteine residues, i.e., cysieine residues not involved in disulfide bonds. The conjugates are hydrolytically stable. Use of cysteine-reactive PEGs allows the development of homogeneous PEG-protein conjugates of defined structure. [0020]
  • Considerable progress has been made in recent years in determining the structures of commercially important protein therapeutics and understanding how they interact with their protein targets, e.g., cell-surface receptors, proteases, etc. This structural information can be used to design PEG-protein conjugates using cysteine-reactive PEGs. Cysteine residues in most proteins participate in disulfide bonds and are not available for PEGylation using cysteine-reactive PEGs. Through in vitro mutagenesis using recombinant DNA techniques, additional cysteine residues can be introduced anywhere into the protein. The added cysteines can be introduced at the beginning of the protein, at the end of the protein, between two amino acids in the protein sequence or, preferably, substituted for an existing amino acid in the protein sequence. The newly added “free” cysteines can serve as sites for the specific attachment of a PEG molecule using cysteine-reactive PEGs. The added cysteine must be exposed on the protein's surface and accessible for PEGylation for this method to be successful. If the site used to introduce an added cysteine site is non-essential for biological activity, then the PEGylated protein will display essentially wild type (normal) in vitro bioactivity. The major technical challenge in PEGylating proteins with cysteine-reactive PEGs is the identification of surface exposed, non-essential regions in the target protein where cysteine residues can be added or substituted for existing amino acids without loss of bioactivity. [0021]
  • Cysteine-added variants of a few human proteins and PEG-polymer conjugates of these proteins have been described. U.S. Pat. No. 5,206,344 describes cysteine-added variants of IL-2. These cysteine-added variants are located within the first 20 amino acids from the amino terminus of the mature IL-2 polypeptide chain. The preferred cysteine variant is at position 3 of the mature polypeptide chain, which corresponds to a threonine residue that is O-glycosylated in the naturally occurring protein. Substitution of cysteine for threonine at position 3 yields an IL-2 variant that can be PEGylated with a cysteine-reactive PEG and retain full in vitro bioactivity (Goodson and Katre, 1990). In contrast, natural IL-2 PEGylated with lysine-reactive PEGs displays reduced in vitro bioactivity (Goodson and Katre, 1990). The effects of cysteine substitutions at other positions in IL-2 were not reported. [0022]
  • U.S. Pat. No. 5,166,322 teaches cysteine added variants of IL-3. This variants are located within the first 14 amino acids from the N-terminus of the mature protein sequence. The patent teaches expression of the proteins in bacteria and covalent modification of the proteins with cysteine-reactive PEGs. No information is provided as to whether the cysteine-added variants and PEG-conjugates of IL-3 are biologically active. Cysteine-added variants at other positions in the polypeptide chain were not reported. [0023]
  • World patent application WO9412219 and PCT application US95/06540 teach cysteine-added variants of insulin-like growth factor-I (IGF-I). IGF-I has a very different structures from GH and is not a member of the GH supergene family (Mott and Campbell, 1995). Cysteine substitutions at many positions in the IGF-I protein are described. Only certain of the cysteine-added variants are biologically active. The preferred site for the cysteine added variant is at amino acid position 69 in the mature protein chain. Cysteine substitutions at positions near the N-terminus of the protein (residues 1-3) yielded IGF-I variants with reduced biological activities and improper disulfide bonds. [0024]
  • World patent application WO9422466 teaches two cysteine-added variants of insulin-like growth factor (IGF) binding protein-1, which has a very different structure than GH and is not a member of the GH supergene family. The two cysteine-added IGF binding protein-1 variants disclosed are located at positions 98 and 101 in the mature protein chain and correspond to serine residues that are phosphorylated in the naturally-occurring protein. [0025]
  • U.S. patent application Ser. No. 07/822296 teaches cysteine added variants of tumor necrosis factor binding protein, which is a soluble, truncated form of the tumor necrosis factor cellular receptor. Tumor necrosis factor binding protein has a very different structure than GH and is not a member of the GH supergene family. [0026]
  • IGF-I, IGF binding protein-1 and tumor necrosis factor binding protein have secondary and tertiary structures that are very different from GH and the proteins are not members of the GH supergene family. Because of this, it is difficult to use the information gained from studies of IGF-I, IGF binding protein-l and tumor necrosis factor binding protein to create cysteine-added variants of members of the GH supergene family. The studies with IL-2 and IL-3 were carried out before the structures of IL-2 and IL-3 were known (McKay 1992; Bazan, 1992) and before it was known that these proteins are members of the GH supergene family. Previous experiments aimed at identifying preferred sites for adding cysteine residues to IL-2 and IL-3 were largely empirical and were performed prior to experiments indicating that members of the GH supergene family possessed similar secondary and tertiary structures. [0027]
  • Based on the structural information now available for members of the GH supergene family, the present invention provides “rules” for determining a priori which regions and amino acid residues in members of the GH supergene family can be used to introduce or substitute cysteine residues without significant loss of biological activity. In contrast to the naturally occurring proteins, these cysteine-added variants of members of the GH supergene family will possess novel properties such as the ability to be covalently modified at defined sites within the polypeptide chain with cysteine-reactive polymers or other types of cysteine-reactive moieties. The covalently modified proteins will be biologically active. [0028]
  • GH is the best-studied member of the GH supergene family. GH is a 22 kDa protein secreted by the pituitary gland. GH stimulates metabolism of bone, cartilage and muscle and is the body's primary hormone for stimulating somatic growth during childhood. Recombinant human GH (rhGH) is used to treat short stature resulting from GH inadequacy and renal failure in children. GH is not glycosylated and can be produced in a fully active form in bacteria. The protein has a short in vivo half-life and must be administered by daily subcutaneous injection for maximum effectiveness (MacGillivray et al., 1996). Recombinant human GH (rhGH) was approved recently for treating cachexia in AIDS patients and is under study for treating cachexia associated with other diseases. [0029]
  • The sequence of human GH is well known (see, e.g., Martial et al. 1979; Goeddel et al. 1979 which are incorporated herein by reference; SEQ ID NO:1). GH is closely related in sequence to prolactin and placental lactogen and these three proteins were considered originally to comprise a small gene family. The primary sequence of GH is highly conserved among animal species (Abdel-Meguid et al., 1987), consistent with the protein's broad species cross-reactivity. The three dimensional folding pattern of porcine GH has been solved by X-ray crystallography (Abdel-Meguid et al., 1987). The protein has a compact globular structure, comprising four amphipathic alpha helical bundles joined by loops. Human GH has a similar structure (de Vos et al., 1992). The four alpha helical regions are termed A-D beginning from the N-terminus of the protein. The loop regions are referred to by the helical regions they join, e.g., the A-B loop joins helical bundles A and B. The A-B and C-D loops are long, whereas the B-C loop is short. GH contains four cysteine residues, all of which participate in disulfide bonds. The disuifide assignments are cysteine53 joined to cysteine165 and cysteine182 joined to cysteine189. [0030]
  • The crystal structure of GH bound to its receptor revealed that GH has two receptor binding sites and binds two receptor molecules (Cunningham et al., 1991; de Vos et al., 1992). The two receptor binding sites are referred to as site I and site II. Site I encompasses the Carboxy (C)-terminal end of helix D and parts of helix A and the A-B loop, whereas site II encompasses the Amino (N)-terminal region of helix A and a portion of helix C. Binding of GH to its receptor occurs sequentially, with site I always binding first. Site II then engages a second GH receptor, resulting in receptor dimerization and activation of the intracellular signaling pathways that lead to cellular responses to GH. A GH mutein in which site II has been mutated (a glycine to arginine mutation at amino acid 120) is able to bind a single GH receptor, but is unable to dimerize GH receptors; this mutein acts as a GH antagonist in vitro, presumably by occupying GH receptor sites without activating intracellular signaling pathways (Fuh et al., 1992). [0031]
  • The roles of particular regions and amino acids in GH receptor binding and intracellular signaling also have been studied using techniques such as mutagenesis, monoclonal antibodies and proteolytic digestion. The first mutagenesis experiments entailed replacing entire domains of GH with similar regions of the closely related protein, prolactin (Cunningham et al., 1989). One finding was that replacement of the B-C loop of GH with that of prolactin did not affect binding of the hybrid GH protein to a soluble form of the human GH receptor, implying that the B-C loop was non-essential for receptor binding. Alanine scanning mutagenesis (replacement of individual amino acids with alanine) identified 14 amino acids that are critical for GH bioactivity (Cunningham and Wells, 1989). These amino acids are located in the helices A, B, C, and D and the A-B loop and correspond to sites I and II identified from the structural studies. Two lysine residues at amino acid positions 41 and 172, K41 and K172, were determined to be critical components of the site I receptor binding site, which explains the decrease in bioactivity observed when K1 72 is acetylated (Teh and Chapman, 1988). Modification of K168 also significantly reduced GH receptor binding and bioactivity (de la Llosa et al., 1985; Martal et al., 1985; Teh and Chapman, 1988). Regions of GH responsible for binding the GH receptor have also been studied using monoclonal antibodies (Cunningham et al., 1989). A series of eight monoclonal antibodies was generated to human GH and analyzed for the ability to neutralize allowed the putative binding site for each monoclonal antibody to be localized within the GH three-dimensional structure. Of interest was that monoclonal antibodies 1 and 8 were unable to displace GH from binding its receptor. The binding sites for these monoclonal antibodies were localized to the B-C loop (monoclonal number 1) and the N-terminal end of the A-B loop (monoclonal number 8). No monoclonals were studied that bound the C-D loop specifically. The monoclonal antibody studies suggest that the B-C loop and N-terminal end of the A-B loop are non-essential for receptor binding. Finally, limited cleavage of GH with trypsin was found to produce a two chain derivative that retained full activity (Mills et al., 1980; Li, 1982). Mapping studies indicated that trypsin cleaved and/or deleted amino acids between positions 134 and 149, which corresponds to the C-D loop. These studies suggest the C-D loop is not involved in receptor binding or GH bioactivity. [0032]
  • Structures of a number of cytokines, including G-CSF (Hill et al., 1993), GM-CSF (Diederichs et al., 1991; Walter et al., 1992), IL-2 (Bazan, 1992; McKay, 1992), IL-4 ( Redfield et al., 1991; Powers et al., 1992), and IL-5 (Milburn et al., 1993) have been determined by X-ray diffraction and NMR studies and show striking conservation with the GH structure, despite a lack of significant primary sequence homology. EPO is considered to be a member of this family based upon modeling and mutagenesis studies (Boissel et al., 1993; Wen et al., 1994). A large number of additional cytokines and growth factors including ciliary neurotrophic factor (CNTF), leukemia inhibitory factor (LIEF), thrombopoietin (TPO), oncostatin M, macrophage colony stimulating factor (M-CSF), IL-3, IL-6, IL-7, IL-9, IL-12, IL-13, IL-15, and alpha, beta, omega, tau and gamma interferon belong to this family (reviewed in Mott and Campbell, 1995; Silvennoinen and Ihle 1996). All of the above cytokines and growth factors are now considered to comprise one large gene family, of which GH is the prototype. [0033]
  • In addition to sharing similar secondary and tertiary structures, members of this family share the property that they must oligomerize cell surface receptors to activate intracellular signaling pathways. Some GH family members, e.g., GH and EPO, bind a single type of receptor and cause it to form homodimers. Other family members, e.g., IL-2, IL-4, and IL-6, bind more than one type of receptor and cause the receptors to form heterodimers or higher order aggregates (Davis et al., 1993; Paonessa et al., 1995; Mott and Campbell, 1995). Mutagenesis studies have shown that, like GH, these other cytokines and growth factors contain multiple receptor binding sites, typically two, and bind their cognate receptors sequentially (Mott and Campbell, 1995; Matthews et al., 1996). Like GH, the primary receptor binding sites for these other family members occur primarily in the four alpha helices and the A-B loop (reviewed in Mott and Campbell, 1995). The specific amino acids in the helical bundles that participate in receptor binding differ amongst the family members (Mott and Campbell, 1995). Most of the cell surface receptors that interact with members of the GH supergene family are structurally related and comprise a second large multi-gene family (Bazan, 1990; Mott and Campbell, 1995; Silvennoinen and Ihle 1996). [0034]
  • A general conclusion reached from mutational studies of various members of the GH supergene family is that the loops joining the alpha helices generally tend to not be involved in receptor binding. In particular the short B-C loop appears to be non-essential for receptor binding in most, if not all, family members. For this reason, the B-C loop is a preferred region for introducing cysteine substitutions in members of the GH supergene family. The A-B loop, the B-C loop, the C-D loop (and D-E loop of interferon/IL-10-like members of the GH superfamily) also are preferred sites for introducing cysteine mutations. Amino acids proximal to helix A and distal to the final helix also tend not to be involved in receptor binding and also are preferred sites for introducing cysteine substitutions. Certain members of the GH family, e.g., EPO, IL-2, IL-3, IL-4, IL-6, G-CSF, GM-CSF, TPO, IL-10, IL-12 p35, IL-13, IL-15 and beta-interferon contain N-linked and O-linked sugars. The glycosylation sites in the proteins occur almost exclusively in the loop regions and not in the alpha helical bundles. Because the loop regions generally are not involved in receptor binding and because they are sites for the covalent attachment of sugar groups, they are preferred sites for introducing cysteine substitutions into the proteins. Amino acids that comprise the N— and O-linked glycosylation sites in the proteins are preferred sites for cysteine substitutions because these amino acids are surface-exposed, the natural protein can tolerate bulky sugar groups attached to the proteins at these sites and the glycosylation sites tend to be located away from the receptor binding sites. [0035]
  • Many additional members of the GH gene family are likely to be discovered in the future. New members of the GH supergene family can be identified through computer-aided secondary and tertiary structure analyses of the predicted protein sequences. Members of the GH supergene family will possess four or five amphipathic helices joined by non-helical amino acids (the loop regions). The proteins may contain a hydrophobic signal sequence at their N-terminus to promote secretion from the cell. Such later discovered members of the GH supergen family also are included within this invention. [0036]
  • The present invention provides “rules” for creating biologically active cysteine-added variants of members of the GH supergene family. These “rules” can be applied to any existing or future member of the GH supergene family. The cysteine-added variants will posses novel properties not shared by the naturally occurring proteins. Most importantly, the cysteine added variants will possess the property that they can be covalently modified with cysteine-reactive polymers or other types of cysteine-reactive moieties to generate biologically active proteins with improved properties such as increased in vivo half-life, increased solubility and improved in vivo efficacy. [0037]
  • Specifically, the present invention provides biologically active cysteine variants of members of the GH supergene family by substituting cysteine residues for non-essential amino acids in the proteins. Preferably, the cysteine residues are substituted for amino acids that comprise the loop regions, for amino acids near the ends of the alpha helices and for amino acids proximal to the first amphipathic helix or distal to the final amphipathic helix of these proteins. Other preferred sites for adding cysteine residues are at the N-terminus or C-terminus of the proteins. Cysteine residues also can be introduced between two amino acids in the disclosed regions of the polypeptide chain. The present invention teaches that N— and O-linked glycosylation sites in the proteins are preferred sites for introducing cysteine substitutions either by substitution for amino acids that make up the sites or, in the case of N-linked sites, introduction of cysteines therein. The glycosylation sites can be serine or threonine residues that are O-glycosylated or asparagine residues that are N-glycosylated. N-linked glycosylation sites have the general structure asparagine-X-serine or threonine (N—X—S/T), where X can be any amino acid. The asparagine residue, the amino acid in the X position and the serine/threonine residue of the N-linked glycosylation site are preferred sites for creating biologically active cysteine-added variants of these proteins. Amino acids immediately surrounding or adjacent to the O-linked and N-linked glycosylation sites (within about 10 residues on either side of the glycosylation site) are preferred sites for introducing cysteine-substitutions. [0038]
  • More generally, certain of the “rules” for identifying preferred sites for creating biologically active cysteine-added protein variants can be applied to any protein, not just proteins that are members of the GH supergene family. Specifically, preferred sites for creating biologically active cysteine variants of proteins (other than IL-2) are O-linked glycosylation sites. Amino acids immediately surrounding the O-linked glycosylation site (within about 10 residues on either side of the glycosylation site) also are preferred sites. N-linked glycosylation sites, and the amino acid residues immediately adjacent on either side of the glycosylation site (within about 10 residues of the N—X—S/T site) also are preferred sites for creating cysteine added protein variants. Amino acids that can be replaced with cysteine without significant loss of biological activity also are preferred sites for creating cysteine-added protein variants. Such non-essential amino acids can be identified by performing cysteine-scanning mutagenesis on the target protein and measuring effects on biological activity. Cysteine-scanning mutagenesis entails adding or substituting cysteine residues for individual amino acids in the polypeptide chain and determining the effect of the cysteine substitution on biological activity. Cysteine scanning mutagenesis is similar to alanine-scanning mutagenesis (Cunningham et al., 1992), except that target amino acids are individually replaced with cysteine rather than alanine residues. [0039]
  • Application of the “rules” to create cysteine-added variants and conjugates of protein antagonists also is contemplated. Excess production of cytokines and growth factors has been implicated in the pathology of many inflammatory conditions such as rheumatoid arthritis, asthma, allergies and wound scarring. Excess production of GH has been implicated as a cause of acromegaly. Certain growth factors and cytokines, e.g., GH and IL-6, have been implicated in proliferation of particular cancers. Many of the growth factors and cytokines implicated in inflammation and cancer are members of the GH supergene family. There is considerable interest in developing protein antagonists of these molecules to treat these diseases. One strategy involves engineering the cytokines and growth factors so that they can bind to, but not oligomerize receptors. This is accomplished by mutagenizing the second receptor binding site (site II) on the molecules. The resulting muteins are able to bind and occupy receptor sites but are incapable of activating intracellular signaling pathways. This strategy has been successfully applied to GH to make a GH antagonist (Cunningham et al., 1992). Similar strategies are being pursued to develop antagonists of other members of the GH supergene family such as IL-2 (Zurawski et al., 1990; Zurawski and Zurawski, 1992), IL-4 (Kruse et al., 1992), IL-5 (Tavernier et al., 1995), GM-CSF (Hercus et al., 1994) and EPO (Matthews et al., 1996). Since the preferred sites for adding cysteine residues to members of the GH supergene family described here lie outside of the receptor binding sites in these proteins, and thus removed from any sites used to create protein antagonists, the cysteine-added variants described herein could be used to generate long-acting versions of protein antagonists. As an example, Cunningham et al. (1992) developed an in vitro GH antagonist by mutating a glycine residue (amino acid 120) to an arginine. This glycine residue is a critical component of the second receptor binding site in GH; when it is replaced with arginine, GH cannot dimerize receptors. The glycine to arginine mutation at position 120 can be introduced into DNA sequences encoding the cysteine-added variants of GH contemplated herein to create a cysteine-added GH antagonist that can be conjugated with cysteine-reactive PEGs or other types of cysteine-reactive moieties. Similarly, amino acid changes in other proteins that turn the proteins from agonists to antagonists could be incorporated into DNA sequences encoding cysteine-added protein variants described herein. Considerable effort is being spent to identify amino acid changes that convert protein agonists to antagonists. Hercus et al.(1994) reported that substituting arginine or lysine for glutamic acid at position 21 in the mature GM-CSF protein converts GM-CSF from an agonist to an antagonist. Tavernier et al.(1995) reported that substituting glutamine for glutamic acid at position 13 of mature IL-5 creates an IL-5 antagonist. [0040]
  • Experimental strategies similar to those described above can be used to create cysteine-added variants (both agonists and antagonists) of members of the GH supergene family derived from various animals. This is possible because the primary amino acid sequences and structures of cytokines and growth factors are largely conserved between human and animal species. For this reason, the “rules” disclosed herein for creating biologically active cysteine-added variants of members of the GH supergene family will be useful for creating biologically active cysteine-added variants of members of the GH supergene family of companion animals (e.g., dogs, cats, horses) and commercial animal (e.g., cow, sheep, pig) species. Conjugation of these cysteine-added variants with cysteine-reactive PEGs will create long-acting versions of these proteins that will benefit the companion animal and commercial farm animal markets. [0041]
  • Proteins that are members of the GH supergene family (hematopoietic cytokines) are provided in Silvennoimem and Ihle (1996). Silvennoimem and lhle (1996) also provide information about the structure and expression of these proteins. DNA sequences, encoded amino acids and in vitro and in vivo bioassays for the proteins described herein are described in Aggarwal and Gutterman (1992; 1996), Aggarwal (1998), and Silvennoimem and lhle (1996). Bioassays for the proteins also are provided in catalogues of various commercial suppliers of these proteins such as R&D Systems, Inc. and Endogen, Inc. [0042]
  • The following examples are provided to demonstrate how these “rules” can be used to create cysteine-added variants of GH, erythropoietin, alpha interferon, beta interferon, G-CSF, GM-CSF and other members of the GH supergene family. The examples are not intended to be limiting, but only exemplary of specific embodiments of the invention. [0043]
  • EXAMPLE 1 Cysteine-Added Variants of GH
  • This example discloses certain amino acids in GH that are non-essential for biological activity and which, when mutated to cysteine residues, will not alter the normal disulfide binding pattern and overall conformation of the molecule. These amino acids are located at the N-terminal end of the A-B loop (amino acids 34-52 of the mature protein sequence; SEQ ID NO: 1; Martial et al 1979; Goeddel et al 1979), the B-C loop (amino acids 97-105 in the mature protein sequence), and the C-D loop (amino acids 130-153 in the mature protein sequence). Also identified as preferred sites for introducing cysteine residues are the first three or last three amino acids in the A, B, C and D helices and the amino acids proximal to helix A and distal to helix D. [0044]
  • DNA sequences encoding wild type GH can be amplified using the polymerase chain reaction technique from commercially available single-stranded cDNA prepared from human pituitaries (ClonTech, San Diego, Calif.) or assembled using overlapping oligonucleotides. Specific mutations can be introduced into the GH sequence using a variety of procedures such as phage techniques (Kunkel et al 1987), PCR mutagenesis techniques (Innis et al 1990; White 1993) mutagenesis kits such as those sold by Stratagene (“Quick-Change Mutagenesis” kit, San Diego, Calif.) or Promega (Gene Editor Kit, Madison Wis.). [0045]
  • Cysteine substitutions can be introduced into any of the amino acids comprising the B-C loop, C-D loop and N-terminal end of the A-B loop or into the first three amino acids of the alpha helical regions that adjoin these regions or in the region proximal to helix A or distal to helix D. Preferred sites for introduction of cysteine residues are: F1, T3, P5, E33, A34, K38, E39, Q40, S43, Q46, N47, P48, Q49, T50, S51, S55, T60, A98, N99, S100, G104, A105, S106, E129, D130, G131, S132, P133, T135, G136, Q137, K140, Q141, T142, S144, K145, D147, T148, N149, S150, H151, N152, D153, S184, E186, G187, S188, and G190. Cysteine residues also can be introduced at the beginning of the mature protein, i.e., proximal to the F1 amino acid, or following the last amino acid in the mature protein, i.e., following F191. If desirable, two or more such mutations can be readily combined in the same protein either by in vitro DNA recombination of cloned mutant genes and/or sequential construction of individual desired mutations. [0046]
  • 1. Cloning the Gene for human Growth Hormone (GH) [0047]
  • The human GH gene was amplified from human pituitary single-stranded cDNA (commercially available from CLONTECH, Inc., Palo Alto, Calif.) using the polymerase chain reaction (PCR) technique and primers BB1 and BB2. The sequence of BB1 is 5′-GGGGGTCGACCATATGTTCCCAACCATTCCCTTATCCAG-3′ (SEQ ID NO: 24). The sequence of BB2 is 5′-GGGGGATCCTCACTAGAAGCCACAGCTGCCCTC-3′ (SEQ ID NO: 25). Primer BB1 was designed to encode an initiator methionine preceding the first amino acid of mature GH, phenylalanine, and SalI and NdeI sites for cloning purposes. The reverse primer, BB2, contains a BamHI site for cloning purposes. The PCR 100 microliter reactions contained 20 pmoles of each oligonucleotide primer, 1× PCR buffer (Perkin-Elmer buffer containing MgCl[0048] 2), 200 micromolar concentration of each of the four nucleotides dA, dC, dG and dT, 2 ng of single-stranded cDNA, 2.5 units of Taq polymerase (Perkin-Elmer) and 2.5 units of Pfu polymerase (Stratagene, Inc). The PCR reaction conditions were 96° C. for 3 minutes, 35 cycles of (95° C., 1 minute; 63° C. for 30 seconds; 72° C. for 1 minute), followed by 10 minutes at 72° C. The thermocycler employed was the Amplitron II Thermal Cycler (Thermolyne). The approximate 600 bp PCR product was digested with SalI and BamHI, gel purified and cloned into similarly digested plasmid pUC19 (commercially available from New England BioLabs, Beverly, Mass.). The ligation mixture was transformed into E. coli strain DH5alpha and transformants selected on LB plates containing ampicillin. Several colonies were grown overnight in LB media and plasmid DNA isolated using miniplasmid DNA isolation kits purchased from Qiagen, Inc (Valencia, Calif.). Clone LB6 was determined to have the correct DNA sequence.
  • For expression in [0049] E. coli, clone LB6 was digested with NdeI and EcoRI, the approximate 600 bp fragment gel-purified, and cloned into plasmid pCYB1 (commercially available from New England BioLabs, Beverly, Mass.) that had been digested with the same enzymes and phosphatased. The ligation mixture was transformed into E. coli DH5alpha and transformants selected on LB ampicillin plates. Plasmid DNA was isolated from several transformants and screened by digestion with NdeI and EcoRI. A correct clone was identified and named pCYB1: wtGH (pBBT120). This plasmid was transformed into E. coli strains JM109 or W3110 (available from New England BioLabs and the American Type Culture Collection).
  • 2. Construction of STII-GH [0050]
  • Wild type GH clone LB6 (pUC19: wild type GH) was used as the template to construct a GH clone containing the [0051] E. coli STII signal sequence (Picken et al. 1983). Because of its length, the STII sequence was added in two sequential PCR reactions. The first reaction used forward primer BB12 and reverse primer BB10. BB10 has the sequence:
    (SEQ ID NO: 28)
    5′CGCGGATCCGATTAGAATCCACAGCTCCCCTC3′.
  • BB12 has the sequence: [0052]
    5′ATCTATGTTCGTTTTCTCTATCGCTACCAACGCTTACGCATTCCCAACCATTCCCTTATCCAG-3′. (SEQ ID NO: 30)
  • The PCR reactions were as described for amplifying wild type GH except that approximately 4 ng of plasmid LB6 was used as the template rather than single-stranded cDNA and the PCR conditions were 96° C. for 3 minutes, 30 cycles of (95° C. for 1 minute; 63° C. for 30 seconds; 72° C. for 1 minute) followed by 72° C. for 10 minutes. The approximate 630 bp PCR product was gel-purified using the Qiaex II Gel Extraction Kit (Qiagen, Inc), diluted 50-fold in water and 2 microliters used as template for the second PCR reaction. The second PCR reaction used reverse primer BB10 and forward primer BB11. BB11 has the sequence: [0053]
    5′CCCCCTCTAGACATATGAAGAAGAACATCGCATTCCTGCTGGCATCTATGTTCGTTTTCTCTATCG-3′. (SEQ ID NO: 29)
  • Primer BB11 contains XbaI and NdeI sites for cloning purposes. PCR conditions were as described for the first reaction. The approximate 660 bp PCR product was digested with XbaI and BamHI, gel-purified and cloned into similarly cut plasmid pCDNA3.1 (+) (Invitrogen, Inc. Carlsbad, Calif.). Clone pCDNA3.1(+)::stII-GH(5C) or “5C” was determined to have the correct DNA sequence. [0054]
  • Clone “5C” was cleaved with NdeI and BarmHI and cloned into similarly cut pBBT108 (a derivative of pUC19 which lacks a Pst I site, this plasmid is described below). A clone with the correct insert was identified following digestion with these enzymes. This clone, designated pBBT111, was digested with NdeI and SalI, the 660 bp fragment containing the stII-GH fusion gene, was gel-purified and closed into the plasmid expression vector pCYB1 (New England BioLabs) that had been digested with the same enzymes and phosphatased. A recombinant plasmid containing the stII-GH insertion was identified by restriction endonuclease digestions. One such isolate was chosen for further studies and was designated pBBT114. This plasmid was transformed into [0055] E. coli strains JM109 or W3110 (available from New England BioLabs and the American Type Culture Collection).
  • 3. Construction of ompA-GH [0056]
  • Wild type GH clone LB6 (pUC19: wild type GH) was used as the template to construct a GH clone containing the [0057] E. coli ompA signal sequence (Movva et al 1980). Because of its length, the ompA sequence was added in two sequential PCR reactions. The first reaction used forward primer BB7:
    5′GCAGTGGCACTGGCTGGTTTCGCTACCGTAGCGCAGGCCTTCCCAACCATTCCCTTATCCAG3′, (SEQ ID NO: 31)
  • and reverse primer BB10: [0058]
    (SEQ ID NO: 28)
    5′CGCGGATCCGATTAGAATCCACAGCTCCCCTC3′.
  • The PCR reactions were as described for amplifying wild type GH except that approximately 4 ng of plasmid LB6 was used as the template rather than single-stranded cDNA and the PCR conditions were 96° C. for 3 minutes, 30 cycles of (95° C. for 1 minute; 63° C. for 30 seconds; 72° C. for 1 minute) followed by 72° C. for 10 minutes. The approximate 630 bp PCR product was gel-purified using the Qiaex II Gel Extraction Kit (Qiagen, Inc), diluted 50-fold in water and 2 microliters used as template for the second PCR reaction. The second PCR reaction used reverse primer BB10 and forward PrimerBB6: [0059]
    5′CCCCGTCGACACATATGAAGAAGACAGCTATCGCGATTGCAGTGGCACTGGCTGGTTTC3′. (SEQ ID NO: 32)
  • PCR conditions were as described for the first reaction. The approximate 660 bp PCR product was gel-purified, digested with Sal I and Bam H1 and cloned into pUC19 (New England BioLabs) which was cut with Sal I and Bam H1 or pCDNA3.1(+) (Invitrogen) which had been cut by Xho I and Bam H1 ( Sal I and Xho I produce compatible single-stranded overhangs). When several clones were sequenced, it was discovered that all pUC19 clones (8/8) contained errors in the region of the ompA sequence. Only one pCDNA3.1(+) clone was sequenced and it contained a sequence ambiguity in the ompA region. In order to generate a correct ompA-GH fusion gene segments of two sequenced clones which contained different errors separated by a convenient restriction site were recombined and cloned into the pUC19-derivative that lacks the Pst I site (see pBBT108 described below). The resulting plasmid, termed pBBT112, carries the ompA-GH fusion gene cloned as an Nde I-Bam H1 fragment into these same sites in pBBT108. This plasmid is designated pBBT112 and is used in PCR-based, site-specific mutagenesis of GH as described below. [0060]
  • 4. Construction of Pst-pUC19 [0061]
  • To facilitate mutagenesis of the cloned GH gene for construction of selected cysteine substitution and insertion mutations a derivative of the plasmid pUC19 (New England BioLabs) lacking a Pst I site was constructed as follows. pUC19 plasmid DNA was digested with Pst I and subsequently treated at 75 deg. C. with PFU DNA Polymerase (Stratagene) using the vendor-supplied reaction buffer supplemented with 200 uM dNTPs. Under these conditions the polymerase will digest the 3′ single-stranded overhang created by Pst I digestion but will not digest into the double-stranded region. The net result will be the deletion of the 4 single-stranded bases which comprise the middle four bases of the Pst I recognition site. The resulting molecule has double-stranded, i.e., “blunt”, ends. Following these enzymatic reactions, the linear monomer was gel-purified using the Qiaex II Gel Extraction Kit (Qiagen, Inc). This purified DNA was treated with T4 DNA Ligase (New England BioLabs) according to the vendor protocols, digested with Pst I, and used to transform [0062] E. coli DH5alpha. Transformants were picked and analyzed by restriction digestion with Pst I and Bam H1. One of the transformants which was not cleaved by Pst I but was cleaved at the nearby Bam H1 site was picked and designated pBBT108.
  • 5. Construction of GH Muteins [0063]
  • GH muteins were generally constructed using site-directed PCR-based mutagenesis as described in PCR Protocols: Current Methods and Applications edited by B. A. White, 1993 Humana Press, Inc., Totowa, N.J. and PCR Protocols: A Guide to Methods and Applications edited by Innis, M. A. et al 1990 Academic Press Inc San Diego, Calif. Typically PCR primer oligonucleotides are designed to incorporate nucleotide changes to the coding sequence of GH that result in substitution of a cysteine residue for an amino acid at a specific position within the protein. Such mutagenic oligonucleotide primers can also be designed to incorporate an additional cysteine residue at the carboxy terminus or amino terminus of the coding sequence of GH. In this latter case one or more additional amino acid residues could also be incorporated at the amino terminal and/or carboxy terminal to the added cysteine residue if that were desirable. Moreover, oligonucleotides can be designed to incorporate cysteine residues as insertion mutations at specific positions within the GH coding sequence if that were desirable. Again, one or more additional amino acids could be inserted along with the cysteine residue and these amino acids could be positioned at the amino terminal and/or carboxy terminal to the cysteine residue. [0064]
  • The cysteine substitution mutation T135C was constructed as follows. The mutagenic reverse oligonucleotide BB28: [0065]
    (SEQ ID NO: 33)
    5′CTGCTTGAAGATCTGCCCACACCGGGGGCTGCCATC3′
  • was designed to change the codon ACT for threonine at amino acid residue 135 to a TGT codon encoding cysteine and to span the nearby Bgl II site. This oligonucleotide was used in PCR along with the forward oligonucleotide BB34 5′GTAGCGCAGGCCTTCCCAACCATT3′ (SEQ ID NO: 34) which anneals to the junction region of the ompA-GH fusion gene and is not mutagenic. The PCR was performed in a 50 ul reaction in 1× PCR buffer (Perkin-Elmer buffer containing 1.5 mM MgCl[0066] 2), 200 micromolar concentration of each of the four nucleotides dA, dC, dG and dT, with each oligonucleotide primer present at 0.5 μM, 5 pg of pBBT112 (described above) as template and 1.25 units of Amplitac DNA Polymerase (Perkin-Elmer) and 0.125 units of PFU DNA Polymerase (Stratagene). Reactions were performed in a Robocycler Gradient 96 thermal cycler (Stratagene). The program used entailed: 95 deg C. for 3 minutes followed by 25 cycles of 95 deg C. for 60 seconds, 45 deg C. or 50 deg C. or 55 deg C. for 75 seconds, 72 deg C. for 60 seconds followed by a hold at 6 deg C. The PCR reactions were analyzed by agarose gel electrophoresis to identify annealing temperatures that gave significant product of the expected size; ˜430 bp. The 45-deg C. reaction was “cleaned up” using the QIAquick PCR Purification Kit (Qiagen), digested with Bgl II and Pst I. The resulting 278 bp BE II-Pst I fragment, which includes the putative T135C mutation, was gel-purified and ligated into pBBT111 the pUC19 derivative carrying the stII-GH fusion gene (described above) which had been digested with Bgl II and Pst I and gel-purified. Transformants from this ligation were initially screened by digestion with Bgl II and Pst I and subsequently one clone was sequenced to confirm the presence of the T135C mutation and the absence of any additional mutations that could potentially be introduced by the PCR reaction or by the synthetic oligonucleotides. The sequenced clone was found to have the correct sequence.
  • The substitution mutation S132C was constructed using the protocol described above for T135C with the following differences: mutagenic reverse oligonucleotide BB29 5′CTGCTTGAAGATCTGCCCAGTCCGGGGGCAGCCATCTTC3′ (SEQ ID NO: 35) was used instead of BB28 and the PCR reaction with annealing temperature of 50 deg C. was used for cloning. One of two clones sequenced was found to have the correct sequence. [0067]
  • The substitution mutation T148C was constructed using an analogous protocol but employing a different cloning strategy. The mutagenic forward oligonucleotide BB30 5′GGGCAGATCTTCAAGCAGACCTACAGCAAGTTCGACTGCAACTCACACAAC3′ (SEQ ID NO: 36) was used in PCR with the non-mutagenic reverse primer BB33 5′CGCGGTACCCGGGATCCGATTAGAATCCACAGCT3′ (SEQ ID NO: 37) which anneals to the most 3′ end of the GH coding sequence and spans the Bam H1 site immediately downstream. PCR was performed as described above with the exception that the annealing temperatures used were 46, 51 and 56 deg C. Following PCR and gel analysis as described above the 46 and 51 deg C. reactions were pooled for cloning. These were digested with Bam H1 and Bgl II, gel-purified and cloned into pBBT111 which had been digested with Bam H1 and Bgl II, treated with Calf intestinal Alkaline Phosphatase (Promega) according to the vendor protocols, and gel-purified. Transformants from this ligation were analyzed by digestion with Bam H1 and Bgl II to identify clones in which the 188 bp Bam H1-Bgl II mutagenic PCR fragment was cloned in the proper orientation. Because Bam H1 and Bgl II generate compatible ends, this cloning step is not orientation specific. Five of six clones tested were shown to be correctly oriented. One of these was sequenced and was shown to contain the desired T148C mutation. The sequence of the remainder of the 188 bp Bam H1-Bgl II mutagenic PCR fragment in this clone was confirmed as correct. [0068]
  • The construction of the substitution mutation S144C was identical to the construction of T148C with the following exceptions. Mutagenic forward oligonucleotide BB31 5′GGGCAGATCTTCAAGCAGACCTACTGCAAGTTCGAC3′ (SEQ ID NO: 38) was used instead of BB30. Two of six clones tested were shown to be correctly oriented. One of these was sequenced and was shown to contain the desired S144C mutation. The sequence of the remainder of the 188 bp Bam H1-Bgl II mutagenic PCR fragment in this clone was confirmed as correct. [0069]
  • A mutation was also constructed that added a cysteine residue to the natural carboxy terminus of GH. The construction of this mutation, termed stp192C, was similar to that of T148C, but employed different oligonucleotide primers. The reverse mutagenic oligonucleotide BB32 5′CGCGGTACCGGATCCTTAGCAGAAGCCACAGCTGCCCTCCAC3′ (SEQ ID NO: 39) which inserts a TGC codon for cysteine between the codon for the carboxy terminal phe residue of GH and the TAA translational stop codon and spans the nearby Bam H1 site was used along with BB34 5′GTAGCGCAGGCCTTCCCAACCATT3′ (SEQ ID NO: 40) which is described above. Following PCR and gel analysis as described above, the 46 deg C. reaction was used for cloning. Three of six clones tested were shown to be correctly oriented. One of these was sequenced and was shown to contain the desired stp192C mutation. The sequence of the remainder of the 188 bp Bam H1-Bal II mutagenic PCR fragment in this clone was confirmed as correct. [0070]
  • Analogous PCR mutagenesis procedures can be used to generate other cysteine mutations. The choice of sequences for mutagenic oligonucleotides will be dictated by the position where the desired cysteine residue is to be placed and the propinquity of useful restriction endonuclease sites. Generally, it is desirable to place the mutation, i.e., the mismatched segment near the middle of the oligonucleotide to enhance the annealing of the oligonucleotide to the template. Appropriate annealing temperatures for any oligonucleotide can be determined empirically. It is also desirable for the mutagenic oligonucleotide to span a unique restriction site so that the PCR product can be cleaved to generate a fragment that can be readily cloned into a suitable vector, e.g., one that can be used to express the mutein or provides convenient restriction sites for excising the mutated gene and readily cloning it into such an expression vector. Sometimes mutation sites and restriction sites are separated by distances that are greater than that which is desirable for synthesis of synthetic oligonucleotides: it is generally desirable to keep such oligonucleotides under 80 bases in length and lengths of 30-40 bases are more preferable. [0071]
  • In instances where this is not possible, genes targeted for mutagenesis could be re-engineered or re-synthesized to incorporate restriction sites at appropriate positions. Alternatively, variations of PCR mutagenesis protocols employed above, such as the so-called “Megaprimer Method” (Barik, S., pp. 277-286 in Methods in Molecular Biology, Vol. 15: PCR Protocols: Current Methods and Applications edited by B. A. White, 1993, Humana Press, Inc., Totowa, N.J.) or “Gene Splicing by Overlap Extension” (Horton, R. Mi., pp. 251-261, in Methods in Molecular Biology, Vol. 15: PCR Protocols: Current Methods and Applications, edited by B. A. White, 1993, Humana Press, Inc., Totowa, N.J.) can also be employed to construct such mutations. [0072]
  • 6. Expression of GH in pCYB1 [0073]
  • To express GH in [0074] E. coli, pBBT120 (GH gene with no leader sequence cloned into the tac expression vector pCYB1) and pBBT114 (GH gene with stII leader sequence cloned into the tac expression vector pCYB1) were transformed into E. coli strains JM109 and W3 110. The parental vector pCYB1 was also transformed into JM109 and W3110. These strains were given the following designations:
  • BOB119: JM109 (pCYB1) [0075]
  • BOB130: W3110 (pCYB1) [0076]
  • BOB129: JM109 (pBBT120) [0077]
  • BOB133: W3110 (pBBT120) [0078]
  • BOB121: JM109 (pBBT114) [0079]
  • BOB132: W3110 (pBBT114) [0080]
  • For expression, strains were grown overnight at 37° C. in Luria Broth (LB) (Sambrook, et al 1989) containing 100 μg/ml ampicillin. These saturated overnight cultures were diluted to ˜0.03 OD at A[0081] 600 in LB containing 100 μg/ml ampicillin and incubated at 37° C. in shake flasks in rotary shaker, typically at 250-300 rpm. ODs were monitored and IPTG was added to a final concentration of 0.5 mM when culture ODs reached ˜0.25 −0.5, typically between 0.3 and 0.4. Cultures were sampled typically at 1, 3, 5 and ˜16 h post-induction. The “˜16 h” time points represented overnight incubation of the cultures and exact times varied from ˜15-20 h. Samples of induced and uninduced cultures were pelleted by centrifugation, resuspended in 1× sample buffer (50 mM Tris-HCl (pH 6.8), 2% sodium lauryl sulfate, 10% glycerol, 0.1% bromphenol blue) with the addition of 1% β-mercaptoethanol when desirable. Samples were boiled for ˜10 minutes or heated to 95° C. for ˜10 minutes. Samples were cooled to room temperature before being loaded onto SDS polyacrylamide gels or were stored at −20° C. if not run immediately. Samples were run on precast 15% polyacrylamide “Ready Gels” (Bio-Rad, Hercules Calif.) using a Ready Gel Cell electrophoresis apparatus (Bio-Rad) according to the vendor protocols. Typically gels were run at 200 volts for ˜35-45 minutes. Gels were stained with Coomassie Blue or were analyzed by Western Blot following electro-blotting. Coomassie staining of whole cell lysates from strains BOB 129, BOB 133, BOB 121 and BOB 132 showed a band of ˜22 kD that co-migrated with purified recombinant human GH standard purchased from Research Diagnostics Inc (Flanders, N.J.). That band was most prominent in induced cultures following overnight induction. However, a band was also observed at that molecular weight in uninduced cultures of these same strains and could also be observed with and without induction in the BOB 119 and BOB 130 control strains that carried the expression vector pCYB1 lacking the GH gene. To clarify this observation, Western Blot analyses were performed on whole cell lysates of induced cultures of strains BOB119, BOB130, BOB129, BOB133, BOB121, and BOB132. Western blots were performed with polyclonal rabbit anti-human GH antiserum purchased from United States Biological; catalogue # G 9000-11 (Swampscott, Mass.) This primary antibody was used at a 1:5000 dilution and its binding was detected with goat anti-rabbit IgG Fc conjugated to alkaline phosphatase, (product # 31341) purchased from Pierce (Rockford, Ill.). This secondary antibody was used at a 1:10,0000 dilution. Alkaline phosphatase activity was detected using the ImmunoPure® Fast Red TR/AS-MX Substrate Kit (Pierce, Rockford Ill.) according to the vendor protocols. The Western Blots clearly demonstrated presence of GH in lysates of induced cultures of BOB129, BOB133, BOB121, and BOB132 at both 3 and 16 h post-induction. In the induced culture of control strains, BOB119 and BOB130, no GH was detected by Western blot at 3 or 16 h post-induction time points.
  • In these preliminary experiments, the highest yields of GH were obtained from BOB132 W3110(pBBT114) in which the GH gene is fused downstream of the stII secretion signal sequence. This strain was tested further to determine if the GH protein was secreted to the periplasm as would be expected. An induced culture of BOB132 was prepared as described above and subjected to osmotic shock according to the procedure of Koshland and Botstein (Cell 20 (1980) pp. 749-760). This procedure ruptures the outer membrane and releases the contents of the periplasm into the surrounding medium. Subsequent centrifugation separates the periplasm contents, present in the supernatant from the remainder of the cell-associated components. In this experiment, the bulk of the GH synthesized by BOB132 was found to be localized to the periplasm. This result is consistent with the finding that the bulk of the total GH is also indistinguishable in size from the purified GH standard, which indicated that the stII signal sequence had been removed. This is indicative of secretion. A larger scale (500 ml) culture of BOB132 was also induced, cultured overnight and subjected to osmotic shock according to the procedure described by Hsiung et al, 1986 (Bio/Technology 4, pp. 991-995). Gel analysis again demonstrated that the bulk of the GH produced was soluble, periplasmic, and indistinguishable in size from the GH standard. This material could also be quantitatively bound to, and eluted from, a Q-Sepharose column using conditions very similar to those described for recombinant human GH by Becker and Hsiung, 1986 (FEBS Lett 204 pp145-150). [0082]
  • 7. Cloning the Human GH Receptor [0083]
  • The human GH receptor was cloned by PCR using forward primer BB3 and reverse primer BB4. BB3 has the sequence: [0084]
    (SEQ ID NO: 26)
    5′-CCCCGGATCCGCCACCATGGATCTCTGGCAGCTGCTGTT-3′.
  • BB4 has the sequence: [0085]
    (SEQ ID NO: 27)
    5′CCCCGTCGACTCTAGAGCTATTAAATACGTAGCTCTTGGG-3′.
  • The template was single-stranded cDNA prepared from human liver (commercially available from CLONTECH Laboratories). Primers BB3 and BB4 contain BamHI and SalI restriction sites, respectively, for cloning purposes. The 100 μl PCR reactions contained 2.5 ng of the single-stranded cDNA and 20 picomoles of each primer in 1× PCR buffer (Perkin-Elmer buffer containing MgCl[0086] 2), 200 micromolar concentration of each of the four nucleotides dA, dC, dG and dT, 2.5 units of Taq polymerase (Perkin-Elmer) and 2.5 units of Pfu polymerase (Stratagene, Inc). The PCR reaction conditions were: 96° C. for 3 minutes, 35 cycles of (95° C., 1 minute; 58° C. for 30 seconds; 72° C. for 2 minutes), followed by 10 minutes at 72° C. The thermocycler employed was the Amplitron II Thermal Cycler (Thermolyne). The approximate 1.9 kb PCR product was digested with BamHI and SalI and ligated with similarly cut plasmid pUC19 ( New England BioLabs). However, none of the transformants obtained from this ligation reaction contained the 1.9 kb PCR fragment. Leung et al (Nature 1987 330 pp537-543) also failed to obtain full-length cDNA clones of the human GH receptor in pUC19. Subsequently the PCR fragment was cloned into a low copy number vector, pACYC184 (New England BioLabs) at the BamHI and SalI sites in this vector. Such clones were obtained at reasonable frequencies but E coli strains carrying the cloned PCR fragment grew poorly, forming tiny and heterogeneous looking colonies, in the presence of chloramphenicol, which is used to select for maintenance of pACYC 184.
  • The PCR fragment was simultaneously cloned into pCDNA3.1 (+) (Invitrogen). The approximate 1.9 kb PCR product was digested with BamHI and SalI and ligated into the BamHI and XhoI cloning sites of pCDNA3.1 (+). Only infrequent transformants from this ligation contained the cloned GH receptor cDNA and all of those were found to contain deletions of segments of the receptor coding sequence. One of these clones was sequenced and found to contain a deletion of 135 bp within the GH receptor coding sequence: the sequence of the rest of the gene was in agreement with that reported by Leung et at (1987). [0087]
  • 8. Cloning the Rabbit GH Receptor [0088]
  • The rabbit GH receptor was cloned by PCR using forward primer BB3 (described above) and reverse primer BB36. BB36 has the sequence [0089]
    (SEQ ID NO: 41)
    5′CCCCGTCGACTCTAGAGCCATTAGATACAAAGCTCTTGGG3′
  • and contains XbaI and Sal I restriction sites for cloning purposes. Rabbit liver poly(A)[0090] + mRNA was purchased from CLONTECH, Inc. and used as the substrate in first strand synthesis of single-stranded cDNA to produce template for PCR amplification. First strand synthesis of single-stranded cDNA was accomplished using a 1 st Strand cDNA Synthesis Kit for RT-PCR (AMV) kit from Boehringer Mannheim Corp (Indianapolis, Ind.) according to the vendor protocols. Parallel first strand cDNA syntheses were performed using random hexamers or BB36 as the primer. Subsequent PCR reactions with the products of the first strand syntheses as templates, were carried out with primers BB3 and BB36 according to the 1 st Strand cDNA Synthesis Kit for RT-PCR (AMV) kit protocol and using 2.5 units of Amplitac DNA Polymerase (Perkin-Elmer) and 0.625 units of Pfu DNA Polymerase (Stratagene). The PCR reaction conditions were 96° C. for 3 minutes, 35 cycles of (95° C., 1 minute; 58° C. for 30 seconds; 72° C. for 2 minutes), followed by 10 minutes at 72° C. The thermocycler employed was the Amplitron II Thermal Cycler (Thermolyne). The expected 1.9 kb PCR product was observed in PCR reactions using random hexamer-primed or BB36 primed cDNA as template. The random hexamer-primed cDNA was used in subsequent cloning experiments. It was digested with Bam H1 and XbaI and run out over a 1.2% agarose gel. This digest generates two fragments (˜365 bp and ˜1600 bp) because the rabbit GH receptor gene contains an internal Bam H1 site. Both fragments were gel-purified. Initially the ˜1600 bp Bam H1-XbaI fragment was cloned into pCDNA3.1(+) which had been digested with these same two enzymes. These clones were readily obtained at reasonable frequencies and showed no evidence of deletions as determined by restriction digests and subsequent sequencing. To generate a full length clone, one of the plasmids containing the 1600 bp Bam H1-Xba I fragment (pCDNA3.1(+):: rab-ghr-2A) was digested with Bam H1, treated with Calf Intestinal Alkaline Phosphatase (Promega) according to the vendor protocols, gel-purified and ligated with the gel purified ˜365 bp Bam H1 fragment that contains the 5′ portion of the rabbit GH receptor gene. Transformants from this ligation were picked and analyzed by restriction digestion and PCR to confirm the presence of the ˜365 bp fragment and to determine its orientation relative to the distal segment of the rabbit GH receptor gene. Three out of four clones analyzed were found to contain the ˜365 bp fragment cloned in the correct orientation for reconstitution of the rabbit GH receptor gene. The lack of complications in the cloning in E coli of the rabbit gene, in contrast to the human gene, is consistent with the results of Leung et al (1987) who also readily obtained full length cDNA clones for the rabbit GH receptor gene but were unable to clone a full length cDNA of the human gene in E coli. The rabbit GH receptor can be employed in assays with human GH as a ligand as it has been shown that the human GH binds the rabbit receptor with high affinity (Leung et al 1987). Plasmids containing the cloned rabbit GH receptor should be sequenced to identify a rabbit GH receptor cDNA with the correct sequence before use.
  • 9. Construction of a Human/Rabbit Chimeric GH Receptor Gene [0091]
  • As an alternative to the rabbit receptor, a chimeric receptor could be constructed which combines the extracellular domain of the human receptor with the transmembrane and cytoplasmic domains of the rabbit receptor. Such a chimeric receptor could be constructed by recombining the human and rabbit genes at the unique Nco I site that is present in each (Leung et al 1987). Such a recombinant, containing the human gene segment located 5′ to, or “upstream” of the Nco I site and the rabbit gene segment 3′ to, or “downstream” of the Nco I site would encode a chimeric receptor of precisely the desired type, having the extracellular domain of the human receptor with the transmembrane and cytoplasmic domains of the rabbit receptor. This would allow analysis of the interaction of GH, GH muteins, and PEGylated GH muteins with the natural receptor binding site but could avoid the necessity of cloning the full length human GH receptor in [0092] E coli.
  • The GH muteins can be expressed in a variety of expression systems such as bacteria, yeast or insect cells. Vectors for expressing GH muteins in these systems are available commercially from a number of suppliers such as Novagen, Inc. (pET15b for expression in [0093] E. coli), New England Biolabs (pC4B1 for expression in E. coli) Invitrogen (pVL1392, pVL1 393, and pMELBAC for expression in insect cells using Baculovirus vectors, Pichia vectors for expression in yeast cells, and pCDNA3 for expression in mammalian cells). Gn has been successfully produced in E. coli as a cytoplasmic protein and as a secreted, periplasmic, protein using the E. coli OmpA or STII signal sequences to promote secretion of the protein into the periplasmic space (Chang et al., 1987; Hsiung et al., 1986). It is preferable that the GH muteins are expressed as secreted proteins so that they do not contain an N-terminal methionine residue, which is not present in the natural human protein. For expression in E. coli, DNA sequences encoding GH or GH muteins can be cloned into E. coli expression vectors such as pET15b that uses the strong T7 promoter or pCYB1 that uses the TAC promoter. Adding IPTG (isopropylthiogalactopyranoside, available form Sigma Chemical Company) to the growth media, can induce expression of the protein. Recombinant GH will be secreted into the periplasmic space from which it can be released by, and subsequently purified, following osmotic shock (Becker and Hsiung, 1986). The protein can be purified further using other chromatographic methods such as ion-exchange, hydrophobic interaction, size-exclusion and reversed phase chromatography, all of which are well known to those of skill in the art (e.g., see Becker and Hsiung, 1986). Protein concentrations can be determined using commercially available protein assay kits such as those sold by BioRad Laboratories (Richmond, Calif.). If the GH proteins are insoluble when expressed in E. coli they can be refolded using procedures well known to those skilled in the art (see Cox et al., 1994 and World patent applications WO9422466 and WO9412219).
  • Alternatively, the proteins can be expressed in insect cells as secreted proteins. The expression plasmid can be modified to contain the GH signal sequence to promote secretion of the protein into the medium. The cDNAs can be cloned into commercially available vectors, e.g., pVL1392 from Invitrogen, Inc., and used to infect insect cells. The GH and GH muteins can be purified from conditioned media using conventional chromatography procedures. Antibodies to rhGH can be used in conjunction with Western blots to localize fractions containing the GH proteins during chromatography. Alternatively, fractions containing GH can be identified using ELISA assays. [0094]
  • The cysteine-added GH variants also can be expressed as intracellular or secreted proteins in eukaryotic cells such as yeast, insect cells or mammalian cells. Vectors for expressing the proteins and methods for performing such experiments are described in catalogues from various commercial supply companies such as Invitrogen, Inc., Stratagene, Inc. and ClonTech, Inc. The GH and GH muteins can be purified using conventional chromatography procedures. [0095]
  • Biological activity of the GH muteins can be measured using a cell line that proliferates in response to GH. Fuh et al. (1992) created a GH-responsive cell line by stably transforming a myeloid leukemia cell line, FDC-P1, with a chimeric receptor comprising the extracellular domain of the rabbit GH receptor fused to the mouse G-CSF receptor. This cell line proliferates in response to GH with a half maximal effective concentration (EC[0096] 50) of 20 picomolar. A similar cell line can be constructed using the published sequences of these receptors and standard molecular biology techniques (Fuh et al., 1992). Alternatively, the extracellular domain of the human GH receptor can be fused to the mouse G-CSF receptor using the published sequences of these receptors and standard molecular biology techniques. Transformed cells expressing the chimeric receptor can be identified by flow cytometry using labeled GH, by the ability of transformed cells to bind radiolabeled GH, or by the ability of transformed cells to proliferate in response to added GH. Purified GH and GH muteins can be tested in cell proliferation assays using cells expressing the chimeric receptor to measure specific activities of the proteins. Cells can be plated in 96-well dishes with various concentrations of GH or GH muteins. After 18 h, cells are treated for 4 hours with 3H-thymidine and harvested for determination of incorporated radioactivity. The EC50 can be determined for each mutein. Assays should be performed at least three times for each mutein using triplicate wells for each data point. GH muteins displaying similar optimal levels of stimulation and EC50 values comparable to or greater than wild type GH are preferable.
  • GH muteins that retain in vitro activity can be PEGylated using a cysteine-reactive 8 kDa PEG-maleimide (or PEG-vinylsulfone) commercially available from Shearwater, Inc. Generally, methods for PEGylating the proteins with these reagents will be similar to those described in world patent applications WO 9412219 and WO 9422466 and PCT application US95/06540, with minor modifications. The recombinant proteins must be partially reduced with dithiothreitol (DTT) in order to achieve optimal PEGylation of the free cysteine. Although the free cysteine is not involved in a disulfide bond, it is relatively unreactive to cysteine-reactive PEGs unless this partial reduction step is performed. The amount of DTT required to partially reduce each mutein can be determined empirically, using a range of DTT concentrations. Typically, a 5-10 fold molar excess of DTT for 30 min at room temperature is sufficient. Partial reduction can be detected by a slight shift in the elution profile of the protein from a reversed-phase column. Care must be taken not to over-reduce the protein and expose additional cysteine residues. Over-reduction can be detected by reversed phase-HPLC (the protein will have a retention time similar to the fully reduced and denatured protein) and by the appearance of GH molecules containing two PEGs (detectable by a molecular weight change on SDS-PAGE). Wild type GH can serve as a control since it should not PEGylate under similar conditions. Excess DTT can be removed by size exclusion chromatography using spin columns. The partially reduced protein can be reacted with various concentrations of PEG-maleimide (PEG: protein molar ratios of 1:1, 5:1,10:1 and 50:1) to determine the optimum ratio of the two reagents. PEGylation of the protein can be monitored by a molecular weight shift using sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE). The lowest amount of PEG that gives significant quantities of mono-pegylated product without giving di-pegylated product will be considered optimum (80% conversion to mono-pegylated product is considered good). Generally, mono-PEGylated protein can be purified from non-PEGylated protein and unreacted PEG by size-exclusion or ion exchange chromatography. The purified PEGylated protein can be tested in the cell proliferation assay described above to determine its specific activity. [0097]
  • The above experiments will allow identification of amino acids in the B-C loop, C-D loop or N-terminal end of the A-B loop in GH that can be changed to cysteine residues, PEGylated and retain in vitro biological activity. These muteins can be tested in animal disease models well known in the art. [0098]
  • Experiments can be performed to confirm that the PEG molecule is attached to the protein at the proper site. This can be accomplished by proteolytic digestion of the protein, purification of the PEG peptide (which will have a large molecular weight) by size exclusion, ion exchange or reversed phase chromatography, followed by amino acid sequencing or mass spectroscopy. The PEG-coupled amino acid will appear as a blank in the amino acid sequencing run. [0099]
  • The pharmacokinetic properties of the PEG-GH proteins can be determined as follows, or as described in world patent application WO9422466. Pairs of rats or mice can receive an intravenous bolus injection of the test proteins. Circulating levels of the proteins are measured over the course of 24 h by removing a small sample of blood from the animals at desired time points. Circulating levels of the test proteins can be quantitated using ELISA assays. Additional experiments can be performed using the subcutaneous route to administer the proteins. Similar experiments should be performed with the non-PEGylated protein to serve as a control. These experiments will reveal whether attachment of a PEG reagent to the protein alters its pharmacokinetic properties. Covalent modification of the protein with PEG should increase the protein's circulating half-life relative to the unPEGylated protein. Larger PEG molecules and/or attachment of multiple PEG molecules should lengthen the circulating half-life longer than smaller PEG molecules. [0100]
  • PEG-GH proteins can be tested in rodent models of growth hormone deficiency (Cox et al., 1994) and cachexia (Tomas et al., 1992; Read et al., 1992) to determine optimum dosing schedules and demonstrate efficacy. These studies can explore different size PEG molecules, e.g., 8 and 20 kDa, and dosing schedules to determine the optimum PEG size and dosing schedule. It is expected that the larger PEG molecule will increase the circulating half-life greater than the smaller PEG molecule and will require less frequent dosing. However, large proteins potentially may have reduced volumes of distribution in vivo; thus, it is possible a 20 kDa PEG attached to GH will limit bioavailability, reducing its efficacy. Rodent models will allow determination of whether this is the case. Once the optimum dosing schedules and PEG sizes are determined, the efficacy of PEG-GH to GH can be compared in the animal models. While all PEG-GH proteins having GH activity are included in the invention, the preferred PEG-GH proteins are those that enhance growth equal or superior to GH, but which can be given less frequently. PEG-GH should be more efficacious than GH when both are administered using the less frequent dosing schedules. [0101]
  • One GH deficiency model that can be used is a hypophysectomized rat. GH stimulates body weight gain and bone and cartilage growth in this model (Cox et al., 1994). Hypophysectomized rats can be purchased from Charles River. Rats can be injected with GH, PEG-GH or placebo and weight gain measured daily over a 10-14 day period. At time of sacrifice, tibial epiphysis width can be determined as a measure of bone growth. Experimental methods for performing these studies are described in Cox et al. (1994). [0102]
  • The efficacy of PEG-GH in rodent cachexia models can be tested in a similar manner. Daily administration of dexamethasone, via osmotic pumps or subcutaneous injection, can be used to induce weight loss (Tomas et al., 1992; Read et al., 1992; PCT patent application US95/06540). [0103]
  • EXAMPLE 2 Cysteine-Added Variants of Erythropoietin
  • This example relates to cysteine-added variants of erythropoietin (EPO). EPO is the hormone primarily responsible for stimulating erythropoiesis or red blood cell formation. EPO acts on immature red blood cell precursors to stimulate their further proliferation and differentiation into mature red blood cells. A commercial pharmaceutical version is available from Amgen, Inc. Human EPO is a 35-39 kDa glycoprotein secreted by the adult kidney. The mature human protein contains 166 amino acids and is heavily glycosylated. The sequence of human EPO (SEQ ID NO: 2) is shown in Lin et al 1985 and Jacobs et al. 1985, which are incorporated herein by reference. The primary sequence of EPO is highly conserved among species (greater than 80% identity; Wen et al., 1994). Sugar groups account for greater than 40% of the protein's mass. Human EPO contains three N-linked glycosylation sites and one O-linked glycosylation site. The N-linked glycosylation sites are conserved in different species whereas the O-linked glycosylation site is not. The extensive glycosylation of EPO has prevented the protein's crystallization, so the X-ray structure of the protein is not known. Human EPO contains four cysteine residues. The disulfide assignments are Cys7 to Cys161 and Cys29 to Cys33. Cys33 is not conserved in mouse EPO, suggesting that the Cys29-Cys33 disulfide bond is not critical to mouse EPO's structure or function. This conclusion also seems to hold for human EPO (Boissel et al., 1993). [0104]
  • The amino acid sequence of EPO is consistent with the protein being a member of the GH supergene family and mutational studies support this view of EPO's structure (Boissel et al., 1993; Wen et al., 1994). A model of the three dimensional structure of EPO, modeled after the GH structure has been proposed (Boissel et al., 1993; Wen et al., 1994). Amino acids in EPO important for receptor binding have been identified through mutagenesis experiments and reside primarily in the N-terminal half of presumptive helix A and the C-terminal half of presumptive helix D (Boissel et al., 1993; Wen et al., 1994; Matthews et al., 1996). Only a single cell surface receptor for EPO has been identified (D'Andrea et al., 1989). It is believed that EPO dimerizes its receptor in much the same way that GH dimerizes its receptor (Matthew's et al., 1996). [0105]
  • Human EPO contains three sites for N-linked glycosylation (asparagine-24,-38 and -83) and one site for O-linked glycosylation (serine-126). The N-linked glycosylation sites are located in the A-B and B-C loops and the O-glycosylation site is located in the C-D loop. The N-linked glycosylation sites are conserved among species whereas the O-linked glycosylation site is adsent in rodent EPO (Wen et al., 1993). A non-O-linked glycosylated human variant containing methionine at position 126 has been described (U.S. Pat. No. 4,703,008). The N-linked sugar groups are heavily branched and contain terminal sialic acid residues (Sasaki et al., 1987; Takeuchi et al., 1988). N-38 and N-83 contain the most highly branched oligosaccharides (Sasaki et al., 1988). [0106]
  • The terminal sialic residues on EPO are critical for the protein's in vivo function because removal of these residues by digestion eliminates in vivo activity (Fukada et al., 1989; Spivak and Hogans, 1989). Loss of activity correlates with faster clearance of the asialated protein from the body. The circulating half-life of the asialated protein in rats is less than ten minutes, in contrast to that of the sialated protein, which is approximately 2 hr (Fukada et al., 1989; Spivak and Hogans, 1989). Thus, in vivo activity of EPO directly correlates with its circulating half-life. [0107]
  • The role of N-linked sugars in EPO's biological activities haS been better defined by mutating, individually and in combination, the three asparagine residues comprising the N-linked glycosylation sites. EPO muteins in which only a single N-linked glycosylation site was mutated, i.e., N24Q, N38Q and N83Q, were secreted from mammalian cells as efficiently as wild type EPO, indicating that N-linked glycosylation at all three sites is not required for protein secretion (N24Q indicates that the asparagine at position 24 is mutated to glutamine). In contrast, EPO muteins in which two or more N-linked glycosylation sites were mutated were secreted less efficiently than wild type EPO from mammalian cells (Yamaguchi et al., 1991; Delorme et al., 1992). Mutagenesis studies found that each of the single N-linked glycosylation site muteins had in vitro biological activities equal to or greater than wild type EPO. Thus, it was concluded that none of the N-linked glycosylation sites is essential for secretion or in vitro biological activity of EPO. In fact, removal of one of the glycosylation sites seemed to improve biological activity (Yamaguchi et al., 1991). [0108]
  • The in vivo biological activity of N-linked glycosylation muteins was studied by two groups. Yamaguchi et al. (1991) concluded that the N24Q and N83Q muteins had in vivo activities greater than wild type EPO, which correlated with their increased in vitro activities. These authors found that the N38Q mutein had decreased in vivo activity, about 60% of wild type EPO. N38 is the most heavily branched of the three N-linked glycosylation sites (Sasaki et al., 1988). Delorme et al. (1992) reported that mutating any of the N-linked glycosylation sites reduced in vivo biological activity by about 50%. Muteins in which two or more glycosylation sites were mutated had decreased in vivo activities in both studies. [0109]
  • The above studies indicate that some N-linked glycosylation is required for in vitro and in vivo activity of EPO. Individually, however, none of the three glycosylation sites is absolutely essential for activity. The N-linked sugars increase the apparent molecular weight of EPO and prolong its circulating half-life, which correlates with bioactivity. Natural EPO and EPO manufactured in mammalian cells have complex N-linked sugars containing galactose and terminal sialic acid residues. The galactose residues are recognized by specific receptors on hepatocytes and promote rapid clearance of EPO from the body unless the galactose residues are masked by the terminal sialic acid residues. [0110]
  • Mutagenesis studies concluded that O-linked glycosylation is not required for in vitro or in vivo function of EPO (Delorme et al., 1992). This is in keeping with the observation that rodent EPO is not O-glycosylated and with the existence of a naturally occurring human EPO variant in which serine-126 is replaced by methionine, with a corresponding lack of O-linked glycosylation. Mutagenesis of serine-126 revealed that certain amino acid changes at this site (to valine, histidine or glutamic acid) yielded EPO muteins with biological activities similar to wild type EPO, whereas other amino acid changes (to alanine or glycine) resulted in EPO molecules with severely reduced activities (Delorme et al., 1992). The effect of changing serine-126 to cysteine was not studied. The in vivo bioactivity of S126V EPO was found to be similar to wild type EPO (Delorme et al., 1992). [0111]
  • The requirement for complex, N-linked carbohydrates containing terminal sialic acid residues for in vivo activity of EPO has limited commercial manufacture of the protein to mammalian cells. The important functions of the sialated N-linked sugars are to prevent protein aggregation, increase protein stability and prolong the circulating half-life of the protein. The terminal sialic acid residues prolong EPO's circulating half-life by masking the underlying galactose residues, which are recognized by specific receptors on hepatocytes and promote clearance of the asialated protein. EPO can be produced in insect cells and is N-glycosylated and fully active in vitro; its activity in vivo has not been reported (Wojchowski et al., 1987). [0112]
  • This example provides for the design of cysteine-added EPO variants and their use in preparing conjugates using cysteine-reactive PEGs and other cysteine-reactive moieties. Certain amino acids in EPO are non-essential for biological activity and can be mutated to cysteine residues without altering the normal disuifide binding patter, and overall conformation of the molecule. These amino acids are located in the A-B loop (amino acids 23-58 of the mature protein sequence), the B-C loop (amino acids 77-89 of the mature protein sequence), the C-D loop (amino acids 108-131 of the mature protein sequence), proximal to helix A (amino acids 1-8) and distal to helix D (amino acids 153-166 of the mature protein sequence). Also contemplated as preferred sites for adding cysteine residues are at the N-terminus or C-terminus of the protein sequence. Preferred sites for cysteine substitutions are the O-linked glycosylation site (serine-126) and the amino acids comprising the three N-linked glycosylation sites (N24, I25, T26, N38, 139, T40, N83, S84, S85). Glycosylation sites are attractive sites for introducing cysteine substitutions and attaching PEG molecules to EPO because (1) these sites are surface exposed; (2) the natural protein can tolerate bulky sugar groups at these positions; (3) the glycosylation sites are located in the putative loop regions and away from the receptor binding site (Wen et al., 1994); and (4) mutagenesis studies indicate these sites (at least individually) are not essential for in vitro or in vivo activity (Yamaguchi et al., 1991; Delorme et al., 1992). As discussed above, the local conformation of the region encompassing the O-glycosylation site region seems to be important for biological activity. Whether a cysteine substitution at position 126 affects biological activity has not been studied. The cysteine-29 to cysteine-33 disulfide bond is not necessary for biological activity of EPO because changing both residues to tyrosine simultaneously yielded a biologically active EPO protein (Boissel et al., 1993; Wen et al., 1994). A “free” cysteine can be created by changing either cysteine-29 or cysteine-33 to another amino acid. Preferred amino acid changes would be to serine or alanine. The remaining “free” cysteine (cysteine-29 or cysteine-33) would be a preferred site for covalently modifying the protein with cysteine-reactive moieties. [0113]
  • Bill et al. (1995) individually substituted cysteine for N24, N38 and N83 and reported that the muteins had greatly reduced in vitro biological activities (less than 20% of wild type activity). Bill et al.(1995) expressed the EPO variants as fusion proteins (fused to glutathionine-S-transferase) in bacteria. One aspect of the present invention is to provide expression systems in which the N24C, N38C and N83C EPO variants will have in vitro biological activities more similar to wild type EPO. [0114]
  • U.S. Pat. No. 4,703,008 contemplates naturally occurring variants of EPO as well as amino acid substitutions that are present in EPO proteins of mammals. Ovine EPO contains a cysteine residue at position 88 of the polypeptide chain. The inventor is unaware of any other naturally occurring human or animal cysteine variants of EPO in which the cysteine residue occurs in the polypeptide regions disclosed herein as being useful for generating cysteine-added EPO variants. U.S. Pat. No. 4,703,008 specifically teaches away from cysteine-added EPO variants by suggesting that expression of EPO might be improved by deleting cysteine residues or substituting naturally occurring cysteine residues with serine or histidine residues. [0115]
  • The mature protein form of EPO can contain 165 or 166 amino acids because of post-translational removal of the C-terminal arginine. Asp-165 is the C-terminus of the 165 amino acid form and Agr-166 is the C-terminal amino acid of the 166 amino acid form. The cysteine substitution and insertion mutations described herein can comprise either the 165 or 166 amino acid forms of mature EPO. [0116]
  • A cDNA encoding EPO can be cloned using the polymerase chain reaction (PCR) technique from the human HepG2 or Hep3B cell lines, which are known to express EPO when treated with hypoxia or cobalt chloride (Wen et al., 1993) and are available from the American Type Culture Collection (ATCC). Cysteine mutations can be introduced into the cDNA by standard phage, plasmid or PCR mutagenesis procedures as described for GH. As described above, the preferred sites for introduction of cysteine substitution mutations are in the A-B loop, the B-C loop, the C-D loop and the region proximal to helix A and distal to helix D. The most preferred sites in these regions are the N— and O-linked glycosylation sites: S126C; N24C; I25C, T26C; N38C; I39C, T40C; N83C, S84C and S85C. Other preferred sites for cysteine substitution mutagenesis are in the A-B loop, the B-C loop and the C-D loop, amino acids surrounding the glycosylation sites and the region of the protein proximal to helix A and distal to helix D (Boissel et al., 1993; Wen et al., 1994). Other preferred sites for cysteine substitutions in these regions are: A1, P2, P3, R4, D8, S9, T27, G28, A30, E31, H32, S34, N36, D43, T44, K45, N47, A50, K52, E55, G57, Q58, G77, Q78, A79, Q86, W88, E89, T107, R110, A111, G113, A114, Q115, K116, E117, A118, S120, P121, P122, D123, A124, A125, A127, A128, T132, K154, T157, G158, E159, A160, T163, G164, D165, and R166. Cysteine residues also can be introduced proximal to the first amino acid of the mature protein, i.e., proximal to A1, or distal to the final amino acid in the mature protein, i.e., distal to D165 or R166. Other variants in which cys-29 or cys-33 have been replaced with other amino acids, preferably serine or alanine, also are provided. [0117]
  • Wild type EPO and EPO muteins can be expressed using insect cells to determine whether the cysteine-added muteins are biologically active. DNAs encoding EPO/EPO muteins can be cloned into the Baculovirus expression vector pVL1392 (available from Invitrogen, Inc. and Sigma Corporation (St. Louis, Mo.) and used to infect insect cells. Recombinant Baculoviruses producing EPO can be identified by Western blots of infected insect cell conditioned media using polyclonal anti-human EPO antiserum (available from R&D Systems). The secreted EPO mutein proteins can be purified by conventional chromatographic procedures well known to those of skill in the art. Protein concentrations can be determined using commercially available protein assay kits or ELISA assay kits (available from R&D Systems and Bio-Rad Laboratories). [0118]
  • Purified EPO and EPO muteins can be tested in cell proliferation assays using EPO-responsive cell lines such as UT7-epo (Wen et al., 1994) or TF1 (available from the ATCC) to measure specific activities of the proteins. Cells can be plated in 96-well microtiter plates with various concentrations of EPO. Assays should be performed in triplicate. After 1-3 days in culture, cell proliferation can be measured by [0119] 3H-thymidine incorporation, as described above for GH. The concentration of protein giving half-maximal stimulation (EC50) can be determined for each mutein. Assays should be performed at least three times for each mutein, with triplicate wells for each data point. EC50 values can be used to compare the relative potencies of the muteins. Alternatively, cell proliferation in response to added EPO muteins can be analyzed using an MTT dye-exclusion assay (Komatsu et al. 1991). Proteins displaying similar optimal levels of stimulation and EC50 values comparable to or greater than wild type EPO are preferable.
  • The above studies confirm identification of amino acid residues in EPO that can be changed to cysteine residues and retain biological activity. Muteins that retain activity can be PEGylated using a cysteine-reactive 8 kDa PEG-maleimide as described above for GH muteins. Wild type EPO should be used as a control since it should not react with the cysteine-reactive PEG under identical partial reduction conditions. The lowest amount of PEG that gives significant quantities of mono-PEGylated product without giving di-PEGylated product should be considered optimum. Mono-PEGylated protein can be purified from non-PEGylated protein and unreacted PEG by size-exclusion or ion exchange chromatography. The purified PEGylated proteins should be tested in the cell proliferation assay described above to determine their bioactivities. [0120]
  • One or more of the PEGylated EPO muteins that retain in vitro bioactivity, are candidates for testing in animal disease models. PEGylation of the protein at the proper amino acid can be determined as described for GH. [0121]
  • In vivo testing of PEGylated EPO muteins expressed using insect cells may require that they be re-engineered for expression in mammalian cells to ensure proper glycosylation. PEG-EPO candidates produced using insect cells can be tested in the animal models described below to determine if they are active in vivo and whether they are as active as PEG-EPO produced using mammalian cell expression systems. For expression in mammalian cells, the EPO muteins can be subdloned into commercially available eukaryotic expression vectors and used to stably transform Chinese Hamster Ovary (CHO) cells (available from the ATCC). Sublines can be screened for EPO expression using ELISA assays. Sufficient quantities of the insect cell- and mammalian cell-produced EPO muteins can be prepared to compare their biological activities in animal anemia models. [0122]
  • In vivo bioactivities of the EPO muteins can be tested using the artificial polycythemia or starved rodent models (Cotes and Bangham., 1961; Goldwasser and Gross., 1975). In the starved rodent model, rats are deprived of food on day one and treated with test samples on days two and three. On day four, rats receive an injection of radioactive iron-59. Approximately 18 h later, rats are anesthetized and blood samples drawn. The percent conversion of labeled iron into red blood cells is then determined. In the artificial polycythemia model, mice are maintained in a closed tank and exposed for several days to hypobaric air. The animals are then brought to normal air pressure. Red blood cell formation is suppressed for several days. On day four or six after return to normal air pressure, mice are injected with erythropoietin or saline. Mice receive one injection per day for one to two days. One day later the animals receive an intravenous injection of labeled iron-59. The mice are euthanized 20 h later and the amount of labeled iron incorporated into red blood cells determined. EPO stimulates red blood cell formation in both models as measured by a dose-dependent increase in labeled iron incorporated into red blood cells. In both models different dosing regimens and different times of injections can be studied to determine if PEG-EPO is biologically active and/or more potent and produces longer acting effects than natural EPO. [0123]
  • EXAMPLE 3 Alpha Interferon
  • Alpha interferon is produced by leukocytes and has antiviral, anti-tumor and immunomodulatory effects. There are at least 20 distinct alpha interferon genes that encode proteins that share 70% or greater amino acid identity. Amino acid sequences of the known alpha interferon species are given in Blatt et al. (1996). A “consensus” interferon that incorporates the most common amino acids into a single polypeptide chain has been described (Blatt et al., 1996). A hybrid alpha interferon protein may be produced by splicing different parts of alpha interferon proteins into a single protein (Horisberger and Di Marco, 1995). Some alpha interferons contain N-linked glycosylation sites in the region proximal to helix A and near the B-C loop (Blatt et al. 1966). The alpha 2 interferon protein (SEQ ID NO: 3) contains four cysteine residues that form two disulfide bonds. The cys1-cys98 disulfide bond (cys1-cys99 in some alpha interferon species such as alpha 1; SEQ ID NO: 4) is not essential for activity. The alpha 2-interferon protein does not contain any N-linked glycosylation sites. The crystal structure of alpha interferon has been determined (Radhakrishnan et al., 1996). [0124]
  • This example provides cysteine added variants in the region proximal to the A helix, distal to the E helix, in the A-B loop, in the B-C loop, in the C-D loop and in the D-E loop. Preferred sites for the introduction of cysteine residues in these regions of the alpha interferon-2 species are: D2, L3, P4, Q5, T6, S8, Q20, R22, K23, S25, F27, S28, K31, D32, R33, D35, G37, F38, Q40, E41, E42, F43, G44, N45, Q46, F47, Q48, K49, A50, N65, S68, T69, K70, D71, S72, S73, A74, A75, D77, E78, T79, Y89, Q90, Q91, N93, N94, E96, A97, Q101, G102, G104, T106, E107, T108, P109, K112, E113, D114, S115, K131, K132, K133, K134, Y135, S136, A139, S152, S154, T155, N156, L157, Q158, E159, S160, L161, R162, S163, K164, E165. Variants in which cysteine residues are introduced proximal to the first amino acid of the mature protein, i.e., proximal to C1, or distal to the final amino acid in the mature protein, i.e., distal to E165 are provided. Other variants in which cys-1 or cys-98 (cys-99 in some alpha interferon species) have been replaced with other amino acids, preferably serine or alanine, also are provided. Other variants in which Cys-1 has been deleted (des Cys-1) also are provided. The cysteine variants may be in the context of any naturally occurring or non-natural alpha interferon sequence, e.g., consensus interferon or interferon protein hybrids. Some naturally occurring alpha interferon species (e.g., alpha interferon-1) contain a naturally occurring “free” cysteine. In such interferon species the naturally occurring free cysteine can be changed to another amino acid, preferably serine or alanine. [0125]
  • This example also provides cysteine variants of other alpha interferon species, including consensus interferon, at equivalent sites in these proteins. The alignment of the alpha interferon-2 species with other known alpha interferon species and consensus interferon is given in Blatt et al. (1996). The crystal structure of alpha interferon-2 has been determined by Rhadhakrishnan et al. (1996). Lydon et al (1985) found that deletion of the first four amino acids from the N-terminus of alpha interferon did not affect biological activity. Valenzuela et al (1985) found that substitution of Phe-47 in alpha interferon-2 with Cys, Tyr or Ser did not alter biological activity of the protein. Cys-1 and Cys-98 have been changed individually to glycine and serine, respectively, without altering biological activity of the protein (DeChiara et al, 1986). [0126]
  • DNA sequences encoding alpha interferon-2 can be amplified from human genomic DNA, since alpha interferon genes do not contain introns (Pestka et al., 1987). The DNA sequence of alpha interferon-2 is given in Goeddel et al. (1980). Alternatively, a cDNA for alpha interferon-2 can be isolated from human lymphoblastoid cell lines that are known to express alpha interferon spontaneously or after exposure to viruses (Goeddell et al., 1980; Pickering et al., 1980). Many of these cell lines are available from the American Type Culture Collection (Rockville, Md.). Specific mutations can be introduced into the alpha interferon sequence using plasmid-based site-directed mutagenesis kits (e.g., Quick-Change Mutagenesis Kit, Stratagene, Inc.), phage mutagenesis strategies or employing PCR mutagenesis as described for GH. [0127]
  • Alpha interferon has been successfully produced in [0128] E. coli as an intracellular protein (Tarnowski et al., 1986; Thatcher and Panayotatos, 1986). Similar procedures can be used to express alpha interferon muteins. Plasmids encoding alpha interferon or alpha interferon muteins can be cloned into an E. coli expression vector such as pET15b (Novagene, Inc.) that uses the strong T7 promoter or pCYB 1 (New England BioLabs, Beverly, Mass.) that uses the TAC promoter. Expression of the protein can be induced by adding IPTG to the growth media.
  • Recombinant alpha interferon expressed in [0129] E. coli is sometimes soluble and sometimes insoluble (Tarnowski et al., 1986; Thatcher and Panayotatos, 1986). Insolubility appears to be related to the degree of overexpression of the protein. Insoluble alpha interferon proteins can be recovered as inclusion bodies and renatured to a fully active conformation following standard oxidative refolding protocols (Thatcher and Panayotatos, 1986; Cox et al., 1994). The alpha interteron proteins can be purified further using other chromatographic methods such as ion-exchange, hydrophobic interaction, size-exclusion and reversed phase resins (Thatcher and Panayotaos, 1986). Protein concentrations can be determined using commercially available protein assay kits (Bio-Rad Laboratories).
  • If [0130] E. coli expression of alpha interferon muteins is not successful, one can express the proteins in insect cells as secreted proteins as described for GH. The proteins can be modified to contain the natural alpha interferon signal sequence (Goeddell et al., 1980) or the honeybee mellitin signal sequence (Invitrogen, Inc.) to promote secretion of the proteins. Alpha interferon and alpha interferon muteins can be purified from conditioned media using conventional chromatography procedures. Antibodies to alpha interferon can be used in conjunction with Western blots to localize fractions containing the alpha interferon proteins during chromatography. Alternatively, fractions containing alpha interferon proteins can be identified using ELISAs.
  • Bioactivities of alpha interferon and alpha interferon muteins can be measured using an in vitro viral plaque reduction assay (Ozes et al., 1992; Lewis, 1995). Human HeLa cells can be plated in 96-well plates and grown to near confluency at 37° C. The cells are then washed and treated for 24 hour with different concentrations of each alpha interferon preparation. Controls should include no alpha interferon and wild type alpha interferon (commercially available from Endogen, Inc., Woburn, Mass.). A virus such as Vesicular stomatitis virus (VSV) or encephalomyocarditis virus (EMCV) is added to the plates and the plates incubated for a further 24-48 hours at 37° C. Additional controls should include samples without virus. When 90% or more of the cells have been killed in the virus-treated, no alpha interferon control wells (determined by visual inspection of the wells), the cell monolayer are stained with crystal violet and absorbance of the wells read using a microplate reader. Alternatively, the cell monolayers can be stained with the dye MTT (Lewis, 1995). Samples should be analyzed in duplicate or triplicate. EC[0131] 50 values (the amount of protein required to inhibit the cytopathic effect of the virus by 50%) can be used to compare the relative potencies of the proteins. Wild type alpha interferon-2 protects cells from the cytopathic effects of VSV and EMCV and has a specific activity of approximately 2×108 units/mg in this assay (Ozes et al., 1992). Alpha interferon muteins displaying EC50 values comparable to wild type Alpha Interferon are preferable.
  • Alpha interferon muteins that retain activity can be PEGylated using procedures is similar to those described for GH. Wild type alpha interferon-2 can serve as a control since it should not PEGylate under similar conditions. The lowest amount of PEG that gives significant quantities of mono-pegylated product without giving di-pegylated product should be considered optimum. Mono-PEGylated protein can be purified from non-PEGylated protein and unreacted PEG by size-exclusion or ion exchange chromatography. The purified PEGylated proteins can be tested in the viral plaque reduction bioassay described above to determine their bioactivities. PEGylated alpha interferon proteins with bioactivities comparable to wild type alpha interferon are preferable. Mapping the PEG attachment site and determination of pharmacokinetic data for the PEGylated protein can be performed as described for GH. [0132]
  • In vivo bioactivities of the PEG-alpha interferon muteins can be tested using tumor xenograft models in nude mice and viral infection models (Balkwill, 1986; Fish et al., 1986). Since PEG-alpha interferon bioactivity may be species-specific, one should confirm activity of the PEGylated protein using appropriate animal cell lines in in vitro virus plaque reduction assays, similar to those described above. Next, one should explore the effects of different dosing regimens and different times of injections to determine if PEG-alpha interferon is more potent and produces longer lasting effects than non-PEGylated alpha interferon. [0133]
  • The novel alpha interferon-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0134]
  • EXAMPLE 4 Beta Interferon
  • Beta interferon is produced by fibroblasts and exhibits antiviral, antitumor and immunomodulatory effects. The single-copy beta interferon gene encodes a preprotein that is cleaved to yield a mature protein of 166 amino acids (Taniguchi et al. 1980; SEQ ID NO: 5). The protein contains three cysteines, one of which, cysteine-17, is “free”, i.e., it does not participate in a disulfide bond. The protein contains one N-linked glycosylation site. The crystal structure of the protein has been determined (Karpusas et al. 1997) [0135]
  • This example provides cysteine-added variants at any of the three amino acids that comprise the N-linked glycosylation sites, i.e., N80C, E81C or T82C. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the E helix, in the A-B loop, in the B-C loop, in the C-D loop and in the D-E loop. Preferred sites for introduction of cysteine residues in these regions are: M1, S2, Y3, N4, L5, Q23, N25, G26, R27, E29, Y30, K33, D34, R35, N37, D39, E42, E43, K45, Q46, L47, Q48, Q49, Q51, K52, E53, A68, F70, R71, Q72, D73, S74, S75, S76, T77, G78, E107, K108, E109, D110, F111, T112, R113, G114, K115, L116, A135, K136, E137, K138, S139, I157, N158, R159, L160, T161, G162, Y163, L164, R165 and N166. Variants in which cysteine residues are introduced proximal to the first amino acid of the mature protein, i.e., proximal to M1, or distal to the final amino acid in the mature protein, i.e., distal to N166 also are provided. [0136]
  • These variants are produced in the context of the natural protein sequence or a variant protein in which the naturally occurring “free” cysteine residue (cysteine-17) has been changed to another amino acid, preferably serine or alanine. [0137]
  • The novel beta interferon-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0138]
  • EXAMPLE 5 Granulocyte Colony-Stimulating Factor (G-CSF)
  • G-CSF is a pleuripotent cytokine that stimulates the proliferation, differentiation and function of granulocytes. The protein is produced by activated monocytes and macrophages. The amino acid sequence of G-CSF (SEQ ID NO: 6) is given in Souza et al. (1986), Nagata et al. (1986a, b) and U.S. Pat. No. 4,810,643 all incorporated herein by reference. The human protein is synthesized as a preprotein of 204 or 207 amino acids that is cleaved to yield mature proteins of 174 or 177 amino acids. The larger form has lower specific activity than the smaller form. The protein contains five cysteines, four of which are involved in disulfide bonds. Cysteine-17 is not involved in a disulfide bond. Substitution of cysteine-17 with serine yields a mutant G-CSF protein that is fully active (U.S. Pat. No. 4,810,643). The protein is O-glycosylated at threonine-133 of the mature protein. [0139]
  • Hill et al., Proceedings of the National Academy of Science vol. 90: 5167-5171 (1993) identifies the amino acids that comprise Helices A-D of G-CSF (see the legend to FIG. 2of this publication). The positions of the four helical regions and the intervening (loop) regions, as well as the regions preceding Helix A and following Helix D, are located within the sequence for human granulocyte colony-stimulating factor at the following positions (positions given relative to SEQ ID NO:6): [0140]
  • Preceding Helix A=residues 1-10; [0141]
  • Helix A=residues 11-39; [0142]
  • A-B loop=residues 40-70; [0143]
  • Helix B=residues 71-91; [0144]
  • B-C loop=residues 92-99; [0145]
  • Helix C=residues 100-123; [0146]
  • C-D loop=residues 124-142; [0147]
  • Helix D=residues 143-17; [0148]
  • Following Helix D=residues 173-174. [0149]
  • This example provides a cysteine-added variant at threonine-133. This example provides other cysteine-added variants in the region proximal to helix A, distal to helix D, in the A-B loop, B-C loop and C-D loop. Preferred sites for introduction of cysteine substitutions in these regions are: T1, P2, L3, G4, P5, A6, S7, S8, L9, P10, Q11, S12, T38, K40, S53, G55, W58, A59, P60, S62, S63, P65, S66, Q67, A68, Q70, A91, E93, G94, S96, E98, G100, G125, M126, A127, A129, Q131, T133, Q134, G135, A136, A139, A141, S142, A143, Q145, Q173 and P174. Variants in which cysteine residues are introduced proximal to the first amino acid of the mature protein, i.e., proximal to T1, or distal to the final amino acid in the mature protein, i.e., distal to P174 are provided. These variants are provided in the context of the natural protein sequence or a variant protein in which the naturally occurring “free” cysteine residue (cysteine-17) has been changed to another amino acid, preferably serine or alanine. [0150]
  • A cDNA encoding human G-CSF can be purchased from R&D Systems (Minneapolis, Minn.) or amplified using PCR from mRNA isolated from human carcinoma cell lines such as 5637 and U87-MG known to express G-CSF constitutively (Park et al., 1989; Nagata, 1994). These cell lines are available from the American Type Culture collection (Rockville, Md.). Specific mutations can be introduced into the G-CSF sequence using plasmid-based site-directed mutagenesis kits (e.g., Quick-Change Mutagenesis Kit, Stratagene, Inc.), phage mutagenesis methods or employing PCR mutagenesis as described for GH. [0151]
  • G-CSF has been successfully produced in [0152] E. coli as an intracellular protein (Souza et al., 1986). One can employ similar procedures to express G-CSF and G-CSF muteins. Plasmids encoding G-CSF or G-CSF muteins can be cloned into an E. coli expression vector such as pET15b (available from Novagen, Inc., Madison, Wis.) that uses the strong T7 promoter or pCYB1 (available from New England BioLabs, Beverly, Mass.) that uses the strong TAC promoter. Expression of the protein can be induced by adding IPTG to the growth media. Recombinant G-CSF expressed in E. coli is insoluble and can be recovered as inclusion bodies. The protein can be renatured to a fully active conformation following standard oxidative refolding protocols (Souza et al., 1986; Lu et al., 1992; Cox et al., 1994). Similar procedures can be used to refold cysteine muteins. The proteins can be purified further using other chromatographic methods such as ion exchange, hydrophobic interaction, size-exclusion and reversed phase resins (Souza et al., 1986; Kuga et al., 1989; Lu et al., 1992). Protein concentrations can be determined using commercially available protein assay kits (Bio-Rad Laboratories).
  • If [0153] E. coli expression of G-CSF or G-CSF muteins is not successful, one can express G-CSF and G-CSF muteins in insect cells as secreted proteins as described for GH. The proteins can be modified to contain the natural G-CSF signal sequence (Souza et al., 1986; Nagata et al., 1986a; Nagata et al, 1986b) or the honeybee mellitin signal sequence (Invitrogen, Inc., Carlsbad, Calif.) to promote secretion of the proteins. G-CSF and G-CSF muteins can be purified from conditioned media using conventional chromatography procedures. Antibodies to G-CSF can be used in conjunction with Western blots to localize fractions containing the G-CSF proteins during chromatography. Alternatively, fractions containing G-CSF proteins can be identified using ELISAs.
  • G-CSF muteins also can be expressed in mammalian cells as described for erythropoietin in Example 2. [0154]
  • Bioactivities of G-CSF and the G-CSF muteins can be measured using an in vitro cell proliferation assay. The mouse NFS-60 cell line and the human AML-193 cell line can be used to measure G-CSF bioactivity (Tsuchiya et al., 1986; Lange et al., 1987; Shirafuji et al., 1989). Both cell lines proliferate in response to human G-CSF. The AML-193 cell line is preferable since it is of human origin, which eliminates the possibility of a false conclusion resulting from species differences. The NFS60 cell lines proliferates in response to G-CSF with a half-maximal effective concentration (EC[0155] 50) of 10-20 picomolar. Purified G-CSF and G-CSF muteins can be tested in cell proliferation assays using these cell lines to determinc specific activities of the proteins, using published methods (Tsuchiya et al., 1986; Lange et al., 1987; Shirafuji et al., 1989). Cells can be plated in 96-well tissue culture dishes with different concentrations of G-CSF or G-CSF muteins. After 1-3 days at 37° C. in a humidified tissue culture incubator, proliferation can be measured by 3H-thymidine incorporation as described for GH. Assays should be performed at least three times for each mutein using triplicate wells for each data point. EC50 values can be used to compare the relative potencies of the muteins. G-CSF muteins displaying similar optimal levels of stimulation and EC50 values comparable to wild type G-CSF are preferable.
  • G-CSF muteins that retain activity can be PEGylated using procedures similar to those described for GH. Wild type G-CSF and ser-17 G-CSF can serve as controls since they should not PEGylate under similar conditions. The lowest amount of PEG that gives significant quantities of mono-PEGylated product without giving di-PEGylated product should be considered optimum. Mono-PEGylated protein can be purified from non-PEGylated protein and unreacted PEG by size-exclusion or ion exchange chromatography. The purified PEGylated proteins can be tested in the cell proliferation assay described above to determine their bioactivities. [0156]
  • The PEG site in the protein can be mapped using procedures similar to those described for GH. Pharmacokinetic data for the PEGylated proteins can be obtained using procedures similar to those described for GH. [0157]
  • Initial studies to demonstrate in vivo efficacy of PEG-G-CSF can be done in normal Sprague-Dawley rats, which can be purchased from Charles River. Groups of rats should receive single subcutaneous or intravenous injections of various doses of G-CSF, PEG-G-CSF or placebo. Animals should be sacrificed at daily intervals for up to a week for determination of neutrophil and total white blood cell counts in peripheral blood. Other blood cell types (platelets and red blood cells) can be measured to demonstrate cell specificity. [0158]
  • The efficacy of PEG-G-CSF can be tested in a rat neutropenia model. Neutropenia can be induced by treatment with cyclophosphamide, which is a commonly used chemotherapeutic agent that is myelosuppressive. G-CSF accelerates recovery of normal neutrophil levels in cyclophosphamide-treated animals (Kubota et al., 1990). Rats receive an injection of cyclophosphamide on day 0 to induce neutropenia. The animals are then be divided into different groups, which will receive subcutaneous injections of G-CSF, PEG-G-CSF or placebo. Neutrophil and total white blood cell counts in peripheral blood should be measured daily until they return to normal levels. Initially one should confirm that G-CSF accelerates recovery from neutropenia when injected daily. Next, one should explore the effects of different dosing regimens and different times of injections to determine if PEG-G-CSF is more potent and produces longer lasting effects than non-PEGylated G-CSF. [0159]
  • The novel GCSF-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0160]
  • EXAMPLE 6 Thrombopoietin (TPO)
  • Thrombopoietin stimulates the development of megakaryocyte precursors of platelets. The amino acid sequence of TPO (SEQ ID NO: 7) is given in Bartley et al.(1994), Foster et al.(l 994), de Sauvage et al.(1994), each incorporated herein by reference. [0161]
  • The protein is synthesized as a 353 amino acid precursor protein that is cleaved to yield a mature protein of 332 amino acids. The N-terminal 154 amino acids have homology with EPO and other members of the GH supergene family. The C-terminal 199 amino acids do not share homology with any other known proteins. The C-terminal region contains six N-linked glycosylation sites and multiple O-linked glycosylation sites (Hoffman et al., 1996). O-linked glycosylation sites also are found in the region proximal to the A helix, in the A-B loop, and at the C-terminus of Helix C (Hoffman et al., 1996). A truncated TPO protein containing only residues 1-195 of the mature protein is fully active in vitro (Bartley et al., 1994). [0162]
  • This example provides cysteine-added variants at any of the amino acids that comprise the N-linked glycosylation sites and the O-linked glycosylation sites. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. [0163]
  • Preferred sites for introduction of cysteine residues are: S1, P2, A3, P4, P5, A6, T37, A43, D45, S47, G49, E50, K52, T53, Q54, E56, E57, T58, A76, A77, R78, G79, Q80, G82, T84, S87, S88, G109, T110, Q111, P113, P114, Q115, G116, R117, T118, T119, A120, H121, K122, G146, G147, S148, T149, A155, T158,T159, A160, S163, T165, S166, T170, N176, R177, T178, S179, G180, E183, T184, N185, F186, T187, A188, S189, A190, T192, T193, G194, S195, N213, Q214, T215, S216, S218, N234, G235, T236, S244, T247, S254, S255, T257, S258, T260, S262, S272, S274, T276, T280, T291, T294, S307, T310, T312, T314, S315, N319, T320, S321, T323, S325, Q326, N327, L328, S329, Q330, E331, and G332. Variants in which cysteine residues are introduced proximal to the first amino acid of the mature protein, i.e., proximal to S1, or distal to the final amino acid in the mature protein, i.e., distal to G332 are provided. The cysteine-added variants are provided in the context of the natural human protein or a variant protein that is truncated between amino acids 147 and the C-terminus of the natural protein, G332. Variants in which cysteine residues are added distal to the final amino acid of a TPO protein that is truncated between amino acids 147 and 332 also are provided. [0164]
  • The novel TPO-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0165]
  • EXAMPLE 7 Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF)
  • GM-CSF stimulates the proliferation and differentiation of various hematopoietic cells, including neutrophil, monocyte, eosinophil, erythroid, and megakaryocyte cell lineages. The amino acid sequence of human GM-CSF (SEQ ID NO: 8) is given in Cantrell et al. (1985) and Lee et al (1985) both incorporated herein by reference. [0166]
  • GM-CSF is produced as a 144 amino acid preprotein that is cleaved to yield a mature 127 amino acid protein. The mature protein has two sites for N-linked glycosylation. One site is located at the C-terminal end of Helix A; the second site is in the A-B loop. [0167]
  • This example provides cysteine-added variants at any of the amino acids that comprise the N-linked glycosylation sites, i.e., N27C, L28C, S29C, N37C, E38C and T39C. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Preferred sites for introduction of cysteine substitutions in these regions are: A1, P2, A3, R4, S5, P6, S7, P8, S9, T10, Q11, R30, D31, T32, A33, A34, E35, E41, S44, E45, D48, Q50, E51, T53, Q64, G65, R67, G68, S69, L70, T71, K72, K74, G75, T91, E93, T94, S95, A97, T98, T102, I117, D120, E123, V125, Q126 and E127. Variants in which cysteine residues are introduced proximal to the first amino acid of the mature protein, i.e., proximal to A1, or distal to the final amino acid in the mature protein, i.e., distal to E127 are provided. [0168]
  • The novel GM-CSF-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0169]
  • EXAMPLE 8 IL-2
  • IL-2 is a T cell growth factor that is synthesized by activated T cells. The protein stimulates clonal expansion of activated T cells. Human IL-2 is synthesized as a 153 amino acid precursor that is cleaved to yield a 133 amino acid mature protein (Tadatsugu et al., 1983; Devos et al., 1983; SEQ ID NO: 9). [0170]
  • The amino acid sequence of IL-2 is set forth in (Tadatsugu et al. 1983; Devos et al. 1983). The mature protein contains three cysteine residues, two of which form a disulfide bond. Cysteine-125 of the mature protein is not involved in a disulfide bond. Replacement of cysteine-125 with serine yields an IL-2 mutein with full biological activity (Wang et al., 1984). The protein is O-glycosylated at threonine-3 of the mature protein chain. [0171]
  • This example provides cysteine-added variants in the last four positions of the D helix, in the region distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. These variants are provided in the context of the natural protein sequence or a variant protein in which the naturally occurring “free” cysteine residue (cysteine-125) has been changed to another amino acid, preferably serine or alanine. Variants in which cysteine residues are introduced proximal to the first amino acid, i.e., A1, or distal to the final amino acid, i.e., T133, of the mature protein, also are provided. [0172]
  • The novel IL-2-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0173]
  • EXAMPLE 9 IL-3
  • IL-3 is produced by activated T cells and stimulates the proliferation and differentiation of pleuripotent hematopoietic stem cells. The amino acid sequence of human IL-3 (SEQ ID NO: 10) is given in Yang et al. (1986); Dorssers et al. (1987) and Otsuka et al. (1988) all incorporated herein by reference. The protein contains two cysteine residues and two N-linked glycosylation sites. Two alleles have been described, resulting in isoforms having serine or proline at amino acid position 8 or the mature protein. [0174]
  • This example provides cysteine-added variants at any of the amino acids that comprise the N-linked glycosylation sites. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which cysteine residues are introduced proximal to the first amino acid or distal to the final amino acid in the mature protein, also are provided. [0175]
  • The novel IL-3-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0176]
  • EXAMPLE 10 IL-4
  • [0177]
  • IL-4 is a pleiotropic cytokine that stimulates the proliferation and differentiation of monocytes and T and B cells. IL-4 has been implicated in the process that leads to B cells secreting IgE, which is believed to play a role in asthma and atopy. The bioactivity of IL-4 is species specific. IL-4 is synthesized as a 153 amino acid precursor protein that is cleaved to yield a mature protein of 129 amino acids. The amino acid sequence of human IL-4 (SEQ ID NO: 11) is given in Yokota et al. (1986) which is incorporated herein by reference. The protein contains six cysteine residues and two N-linked glycosylation sites. The glycosylation sites are located in the A-B and C-D loops. [0178]
  • This example provides cysteine-added variants at any of the amino acids that comprise the N-linked glycosylation sites. This, example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which cysteine residues are introduced proximal to the first amino acid, H1, or distal to the final amino acid, S129, of the mature protein are provided. [0179]
  • The novel IL-4-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0180]
  • EXAMPLE 11 IL-5
  • IL-5 is a differentiation and activation factor for eosinophils. The amino acid sequence of human IL-5 (SEQ ID NO: 12) is given in Yokota et al. (1987) which is incorporated herein by reference. The mature protein contains 115 amino acids and exists in solution as a disulfide-linked homodimer. The protein contains both O-linked and N-linked glycosylation sites. [0181]
  • This example provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid in the mature protein are provided. [0182]
  • The novel IL-5-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0183]
  • EXAMPLE 12 IL-6
  • IL-6 stimulates the proliferation and differentiation of many cell types. The amino acid sequence of human IL-6 (SEQ ID NO: 13) is given in Hirano et al. (1986) which is incorporated herein by reference. IL-6 is synthesized as a 212 amino acid preprotein that is cleaved to generate a 184 amino acid mature protein. The mature protein contains two sites for N-linked glycosylation and one site for O-glycosylation, at T137, T138, T142 or T143. [0184]
  • This example provides cysteine-added variants at any of the amino acids that comprise the N-linked glycosylation sites and at the O-linked glycosylation site. Also provided are cysteine-added variants in the region proximal to the A helix, distal to the D nelix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided. [0185]
  • The novel IL-6-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0186]
  • EXAMPLE 13 IL-7
  • IL-7 stimulates proliferation of immature B cells and acts on mature T cells. The amino acid sequence of human IL-7 (SEQ ID NO: 14) is given in Goodwin et al. (1989) which is incorporated herein by reference. The protein is synthesized as a 177 amino acid preprotein that is cleaved to yield a 152 amino acid mature protein that contains three sites for N-linked glycosylation. [0187]
  • This example provides cysteine-added variants at any of the amino acids that comprise the N-linked glycosylation sites. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. [0188]
  • The novel IL-7-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided. [0189]
  • EXAMPLE 14 IL-9
  • IL-9 is a pleiotropic cytokine that acts on many cell types in the lymphoid, myeloid and mast cell lineages. IL-9 stimulates the proliferation of activated T cells and cytotoxic T lymphocytes, stimulates proliferation of mast cell precursors and synergizes with erythropoietin in stimulating immature red blood cell precursors. The amino acid sequence of human IL-9 (SEQ ID NO: 15) is given in Yang et al. (1989) which is incorporated herein by reference. IL-9 is synthesized as a precursor protein of 144 amino acids that is cleaved to yield a mature protein of 126 amino acids. The protein contains four potential N-linked glycosylation sites. [0190]
  • This example provides cysteine-added variants at any of the three amino acids that comprise the N-linked glycosylation sites. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided. [0191]
  • The novel IL-9-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0192]
  • EXAMPLE 15 IL-10
  • The amino acid sequence of human IL-10 (SEQ ID NO: 16) is given in Vieira et al. (1991) which is incorporated herein by reference. IL-10 is synthesized as a 178 amino acid precursor protein that is cleaved to yield a mature protein of 160 amino acids. IL-10 can function to activate or suppress the immune system. The protein shares structural homology with the interferons, i.e., it contains five amphipathic helices. The protein contains one N-linked glycosylation site. [0193]
  • This example provides cysteine-added variants at any of the three amino acids comprising the N-linked glycosylation site. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the E helix, in the A-B loop, in the B-C loop, in the C-D loop and in the D-E loop. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided. [0194]
  • The novel IL-10-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0195]
  • EXAMPLE 16 IL-11
  • IL-11 is a pleiotropic cytokine that stimulates hematopoiesis, lymphopoeisis and acute phase responses. IL-11 shares many biological effects with IL-6. The amino acid sequence of human IL-11 (SEQ ID NO: 17) is given in Kawashima et al. (1991) and Paul et al. (1990) both incorporated herein by reference. IL-11 is synthesized as a precursor protein of 199 amino acids that is cleaved to yield a mature protein of 178 amino acids. There are no N-linked glycosylation sites in the protein. [0196]
  • This example provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided. [0197]
  • The novel IL-11-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0198]
  • EXAMPLE 17 IL-12 p35
  • IL-12 stimulates proliferation and differentiation of NK cells and cytotoxic T lymphocytes. IL-12 exists as a heterodimer of a p35 subunit and a p40 subunit. The p35 subunit is a member of the GH supergene family. The amino acid sequence of the p35 subunit (SEQ ID NO: 18) is given in Gubler et al. (1991) and Wolf et al. (1991) both incorporated herein by reference. p35 is synthesized as a precursor protein of 197 amino acids and is cleaved to yield a mature protein of 175 amino acids. The protein contains 7 cysteine residues and three potential N-linked glycosylation sites. [0199]
  • This example provides cysteine-added variants at any of the three amino acids that comprise the three N-linked glycosylation sites. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. These variants are provided in the context of the natural protein sequence or a variant protein in which the naturally occurring “free” cysteine residue has been changed to another amino acid, preferably serine or alanine. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided. [0200]
  • The novel IL-12 p35-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0201]
  • EXAMPLE 18 IL-13
  • IL-13 shares many biological properties with IL-4. The amino acid sequence of human IL-13 (SEQ ID NO: 19) is given in McKenzie et al. (1993) and Minty et al. (1993) both incorporated herein by reference. The protein is synthesized as a 132 amino acid precursor protein that is cleaved to yield a mature protein of 112 amino acids. The mature protein contains 5 cysteine residues and multiple N-linked glycosylation sites. A variant in which glutamine at position 78 is deleted due to alternative mRNA splicing has been described (McKenzie et al 1993) [0202]
  • This example provides cysteine-added variants at any of the three amino acids comprising the N-linked glycosylation sites. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. These variants are provided in the context of the natural protein sequence or a variant sequence in which the pre-existing “free” cysteine has been changed to another amino acid, preferably to alanine or serine. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided. [0203]
  • The novel IL-13-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0204]
  • EXAMPLE 19 IL-15
  • IL-15 stimulates the proliferation and differentiation of T cells, NK cells, LAK cells and Tumor Infiltrating Lymphocytes. IL-15 may be useful for treating cancer and viral infections. The sequence of IL-15 (SEQ ID NO: 20) is given in Anderson et al. (1995) which is incorporated herein by reference. IL-15 contains two N-linked glycosylation sites, which are located in the C-D loop and C-terminal end of the D helix. IL-15 encodes a 162 amino acid preprotein that is cleaved to generate a mature 114 amino acid protein. [0205]
  • This example provides cysteine-added variants at any of the three amino acids comprising the N-linked glycosylation sites in the C-D loop or C-terminal end of the D helix. This example also provides cysteine-added variants proximal to helix A, in the A-B loop, the B-C loop, the C-D loop or distal to helix D. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided. [0206]
  • The novel IL-15-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0207]
  • EXAMPLE 20 Macrophage Colony Stimulating Factor (M-CSF)
  • M-CSF regulates the growth, differentiation and function of monocytes. The protein is a disulfide-linked homodimer. Multiple molecular weight species of M-CSF, which arise from differential mRNA splicing, have been described. The amino acid sequence of human M-CSF and its various processed forms are given in Kawasaki et al (1985), Wong et al. (1987) and Cerretti et al (1988) which are incorporated herein by reference. Cysteine-added variants can be produced following the general teachings of this application and in accordance with the examples set forth herein. [0208]
  • The novel MCSF-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0209]
  • EXAMPLE 21 Oncostatin M
  • Oncostatin M is a multifunctional cytokine that affects the growth and differentiation of many cell types. The amino acid sequence of human oncostatin M (SEQ ID NO: 21) is given in Malik et al. (1989) which is incorporated herein by reference. Oncostatin M is produced by activated monocytes and T lymphocytes. Oncostatin M is synthesized as a 252 amino acid preprotein that is cleaved sequentially to yield a 227 amino acid protein and then a 196 amino acid protein (Linsley et al., 1990). The mature protein contains O-linked glycosylation sites and two N-linked glycosylation sites. The protein is O-glycosylated at T160, T162 and S165. The mature protein contains five cysteine residues. [0210]
  • This example provides cysteine-added variants at either of the three amino acids comprising the N-linked glycosylation sites or at the amino acids that comprise the O-linked glycosylation sites. This example also provides cysteine-added variants proximal to helix A, in the A-B loop, in the B-C loop, in the C-D loop or distal to helix D. These variants are provided in the context of the natural protein sequence or a variant sequence in which the pre-existing “free” cysteine has been changed to another amino acid, preferably to alanine or serine. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided. [0211]
  • The novel Oncostatin M-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0212]
  • EXAMPLE 22 Ciliary Neurotrophic Factor (CNTF)
  • The amino acid sequence of human CNTF (SEQ ID NO: 22) is given in Lam et al. (1991) which is incorporated herein by reference. CNTF is a 200 amino acid protein that contains no glycosylation sites or signal sequence for secretion. The protein contains one cysteine residue. CNTF functions as a survival factor for nerve cells. [0213]
  • This example provides cysteine-added variants proximal to helix A, in the A-B loop, the B-C loop, the C-D loop or distal to helix D. These variants are provided in the context of the natural protein sequence or a variant sequence in which the pre-existing “free” cysteine has been changed to another amino acid, preferably to alanine or serine. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided. [0214]
  • The novel CNTF-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0215]
  • EXAMPLE 23 Leukemia Inhibitory Factor (LIF)
  • The amino acid sequence of LIF (SEQ ID NO: 23) is given in Moreau et al. (1988) and Gough et al. (1988) both incorporated herein by reference. The human gene encodes a 202 amino acid precursor that is cleaved to yield a mature protein of 180 amino acids. The protein contains six cysteine residues, all of which participate in disulfide bonds. The protein contains multiple O— and N-linked glycosylation sites. The crystal structure of the protein was determined by Robinson et al. (1994). The protein affects the growth and differentiation of many cell types. [0216]
  • This example provides cysteine-added variants at any of the three amino acids comprising the N-linked glycosylation sites or the O-linked glycosylation site. Also provided are cysteine-added variants proximal to helix A, in the A-B loop, the B-C loop, the C-D loop or distal to helix D. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided. [0217]
  • The novel LIF-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. [0218]
  • All of the documents cited herein are incorporated herein by reference. [0219]
  • The protein analogues disclosed herein can be used for the known therapeutic uses of the native proteins in essentially the same forms and doses all well known in the art. [0220]
  • While the exemplary preferred embodiments of the present invention are described herein with particularity, those having ordinary skill in the art will recognize changes, modifications, additions, and applications other than those specifically described herein, and may adapt the preferred embodiments and methods without departing from the spirit of this invention. [0221]
  • References: [0222]
  • Abdel-Meguid, S. S., Shieh, h.-S., Smith W. W., Dayringer, H. E., Violand, B. N. and Bentle, L. A. (1987) Proc. Natl. Acad. Sci. USA 84: 6434-6437. [0223]
  • Abuchowski, A., Kazo, G. M., Verhoest, C. R., van Es, T., Kafkewitz, D., Nucci, M. L., Viau, A. T. and Davis, F. F. (1984) Cancer Biochem. Biophys. 7: 175-186. [0224]
  • Aggarwal, B. B. (1998) Human Cytokines. Handbook for Basic and Clinical Research. Volume III. Blackwell Science, Malden, Mass. [0225]
  • Aggarwal, B. B. and Gutterman, J. U. (1992) Human Cytokines. Handbook for Basic and Clinical Research. Volume I. Blackwell Scientific Publications, Cambridge, Mass. [0226]
  • Aggarwal, B. B. and Gutterman, J. U. (1996) Human Cytokines. Handbook for Basic and Clinical Research. Volume II. Blackwell Science, Cambridge, Mass. [0227]
  • Anderson, D. M., Johnson, L., Glaccum, M. B., Copeland, N. G., Gilbert, D. J., Jenkins, N. A., Valentine, V., Kirstein, M. N., Shapiro, D. N., Morris, S. W., Grabstein, K. and cosman, D. (1995) Genomics 25: 701-706. [0228]
  • Balkwill, F. R. (1986) Methods Enzymology 119: 649-657. [0229]
  • Bartley, T. D., Bogenberger, J. et al., (1994) Cell 77: 1117-1124. [0230]
  • Bazan, F. (1991) Immunology Today 11: 350-354. [0231]
  • Bazan, J. F. (1992) Science 257: 410-411. [0232]
  • Becker, G. W. and Hsiung, H. M. (1986) FEBS Letters 204: 145-150. [0233]
  • Bill, R. M., Winter, P. C., McHale, C. M., Hodges, V. M., Elder, G. E., Caley, J., Flitsch, S. L., Bicknell, R. Lappin, T. R. J. (1995) Biochem. Biophys. Acta 1261: 35-43. [0234]
  • Bittorf, T., Jaster, R., Brock, J. (1993) FEBS Letters 336: 133-136. [0235]
  • Blatt, L. M., Davis, J. M., Klein, S. B. and Taylor, M. W. (1996) Journal of Interferon and cytokine Research 16: 489-499. [0236]
  • Boissel, J.-P., Lee, W.-R., Presnell, S. R., Cohen, F. E. and Bunn, H. F. (1993) J. Biol. Chem. 268: 15983-15993. [0237]
  • Butler, C. A. (1996) Lehman Brothers Technical Report “A Paradigm for Success”. [0238]
  • Cantrell, M. A., Anderson, D., Cerretti, D. P., Price, V., McKereghan, K., Tushinski, R. J., Mochizuki, D. Y., Larsen, A., Grabstein, K., Gillis, S. and Cosman, D. Proc. Natl. Acad. Sci. USA 82: 6250-6254. [0239]
  • Cerretti, D. P. et al. (1988) Molecular Immunology 25: 761. [0240]
  • Chang, C. N., Rey, B., Bochner, B., Heyneker, H. and Gray, G. (1987) Gene: 189-196. [0241]
  • Cotes, P. M. and Bangham, D. R. (1961) Nature 191: 1065-1067 Cox, G. N., McDermott, M. J., Merkel, E., Stroh, C. A., Ko, S. C., Squires, C. H., Gleason, T. M. and Russell, D. (1994) Endocrinology 135: 1913-1920. [0242]
  • Cunningham, B. C. and Wells, J. A. (1989) Science 244: 1081-1085. [0243]
  • Cunningham, B. C., Jhurani, P., Ng, P. and Wells, J. A. (1989) Science 243: 1330-1336. [0244]
  • Cunningham, B. C., Ultsch, M., de Vos, A. M., Mulkerrin, M. G., Clauser, K. R. and Wells, J. A. (1991) Science 254: 821-825. [0245]
  • D'Andrea, A. D., Lodish, H. F. and Wong, G. G. (1989) Cell 57: 277-285. [0246]
  • Davis, S., Aldrich, T. H., Stahl, N., Pan, L., Taga, T., Kishimoto, T., Ip, N. Y. and Yancopoulus, G. D. (1993) Science 260: 1805-1808. [0247]
  • DeChiara, T. M., Erlitz, F. and Tamowski,k, S. J. (1986) Methods Enzymology, 119: 403-15. de la Llosa, P., Chene, N. and Martal, J. (1985) FEBS Letts. 191: 211-215 Delorme, E., Lorenzini, T., Giffin, J., Martin, F., Jacobsen, F., Boone, T., Elliot, S. (1992) Biochemistry 31: 9871-9876. [0248]
  • de Sauvage, F. J., Hass, P. E., Spencer, S. D. et al. (1994) Nature 369: 533-538. [0249]
  • de Vos, A. M., Ultsch, M. and Kossiakoff, A. A. (1992) Science 255: 306-312. [0250]
  • Devos, R., Plaetinck, G., Cheroutre, H., Simons, G., Degrave, W., Tavernier, J., Remaut, E. and Fiers, W. (1983) Nucleic Acids Research 11: 4307. [0251]
  • Diederichs, K., Boone, T. and Karplus, A. (1991) Science 154: 1779-1782. [0252]
  • Dorssers, L., Burger, H., Bot, F., Delwel, R., Van Kessel, A. H. M. G., Lowenberg, B. and Wagemaker, G. (1987) 55: 115-124. [0253]
  • Dube, S., Fisher, J. W., Powell, J. S. (1988) J. Biol. Chem. 263: 17526-17521. [0254]
  • Fish, E. N., Banerjee, K., Levine, H. L. and Stebbing, N. (1986) Antimicrob. Agents Chemother. 30: 52-56. [0255]
  • Foster, D. C., Sprecher, C. A., Grant, F. J., Kramer, J. M. et al. (1994) Proc. Natl. Acad. Sci USA 91: 13023-13027. [0256]
  • Fukuda, M. N., Sasaki, H., Fukuda, M. (1989) Blood 73: 84-89. [0257]
  • Goeddel, D. V., Heyneker, H. L., Hozumi, T. et al., (1979) Nature 281: 544-548. [0258]
  • Goeddel, D. V., Yelverton, E., Ullrich, A., Heynecker, H. L., Miozzari, G., Holmes, W., Seeburg, P. H., Dull, T., Mat, L., Stebbing, N., Crea, R., Maeda, S., McCandliss, R., Sloma, A., Tabor, J. M., Gross, M., Familletti, P. C. and Pestka, S. (1980) Nature 287: 411-416. [0259]
  • Goldwasser, E. and Gross, M. (1975) Methods Enzymology 37: 109-121. [0260]
  • Goodson, R. J. and Katre, N. V. (1990) Biotechnology 8: 343-346. [0261]
  • Goodwin, R. G., Lupton, S., Schmierer, A., Hjerrild, K. J., Jerzy, R., Clevenger, W., Gillis, S., Cosman, D. and Namen, A. E. (1989) Proc. Natl. Acad. Sci. USA 86: 302-306. [0262]
  • Gough, N. M. et al., (1988) Proc. Natl. Acad. Sci USA 85: 2623-2627. [0263]
  • Gubler, U., Chua, A. O., Schoenhaut, D. S., Dwyer, C. M., McComas, W., Motyka, R., Nabavi, N., Wolitzky, A. G., Quinn, P. M., Familletti, P. C. and Gately, M. K. (1991) Proc. Natl Acad. Sci. USA 88: 4143-4147. [0264]
  • Hershfield, M. S., Buckley, R. H., Greenberg, M. L. et al., (1987) N. Engl. J. Medicine 316: 589-596. [0265]
  • Hill, C. P., Osslund, T. D. and Eisenberg, D. (1993) Proc. Natl. Acad. Sci. USA 90:5167-5171. [0266]
  • Hoffman, R. C., Andersen, H., Walker, K., Krakover, J. D., Patel, S., Stamm, M. R. and Osborn, S. G. (1996) Biochemistry 35: 14849-14861. [0267]
  • Hsiung, H. M., Mayne, N. G. and Becker, G. W. (1986) Biotechnology 4: 991-995. [0268]
  • Hercus, T. R., Bagley, C. J., Cambareri, B., Dottore, M., Woodcock, J. M., Vadas, M. A., Shannon, M. F., and Lopez, A. F. (1994) Proc. Natl. Acad. Sci. USA 91: 5838-5842. [0269]
  • Hirano, T., Yasukawa, K., Harada, H., Taga, T., Watanabe, Y., Matsuda, T., Kashiwamura, S.-I., Nakajima, K., Koyama, K., Iwamatsu, A., Tsunasawa, S., Sakiyama, F., Matsui, H., Takahara, Y., Taniguchi, T. and Kishimoto, T. (1986) Nature 324: 73-76. [0270]
  • Horisberger, M. A. and Di Marco, S. (1995) Pharmac. Ther. 66: 507-534. [0271]
  • Innis, M. A., Gelfand, D. H., Sninsky, J. J. and White, T. J. (1990) PCR Protocols: A Guide to Methods and Applications. Academic Press, San Diego, Calif. [0272]
  • Jacobs, K., Shoemaker, C., Rudersdorf, R. et al., (1985) Nature 313: 806-810. [0273]
  • Karpusas, M., Nolte, M., Benton, C. B., Meier, W., Lipscomb, W. N. and Goelz, S. (1997) Proc. Natl. Acad. Sci. USA 94: 11813-11818. [0274]
  • Katre, N. V. (1990) J. Immunology 144: 209-213. [0275]
  • Katre, N. V., Knauf, M. J. and Laird, W. J. (1987) Proc. Natl. Acad. Sci. USA 84: 1487-1491. [0276]
  • Kawasaki, E. S. et al., (1985) Science 230: 291. [0277]
  • Kawashima, I., Ohsumi, J., Mita-Honjo, K., Shimoda-Takano, Ishikawa, H., Sakakibara, S., Miyadai, K. and Takiguchi, Y. (1991) FEBS Letts. 283: 199-202. [0278]
  • Kingsley, D. M. (1994) Genes Dev. 8: 133-146. [0279]
  • Komatsu, N., Nakauchi, H., Miwa, A., Ishihara, T., Eguguchi, M. et al., (1991) Cancer Research 51, 341-348. [0280]
  • Kruse, N., Tony, H.-P. and Sebald, W. (1992) EMBO J. 11: 3237-3244. [0281]
  • Lam, A., Fuller, F., Miller, J., Kloss, J., Manthorpe, M., Varon, S. and Cordell, B. (1991) Gene 102: 271-276. [0282]
  • Kubota, N., Orita, T., Hattori, K., Oh-eda, M., Ochi, N. and Yamazaki, T. (1990) J. Biochem. 107: 486492. [0283]
  • Kuga, T., Komatsu, Y., Yamasaki, M., De4kine, S., Miyaji, H., Nishi, T., Sato, M., Yokoo, Y., Asano, M., Okabe, M., Morimoto, M. and Itoh, S. (1989) Bioch. Biophys. Res. Comm. 159:103-111. [0284]
  • Kunkel, T. A., Roberts, J. D. and Zakour, R. A. (1987) Methods in Enzymology 154: 367-382. [0285]
  • Lange, B., Valtieri, M., Santoli, D., Caracciolo, D., Mavilio, F., Gemperlein, I., Griffin, C., Emanuel, B., Finan, J., Nowell, P. and Rovera, G. (1987) Blood 70: 192-199. [0286]
  • Lee, F., Yokota, T., Otsuka, T., Gemmell, L., Larson, N., Luh, J., Arai, K.-I. and Rennick, D. (1985) Proc. Natl. Acad. Sci. USA 82: 4360-4364. [0287]
  • Lewis, J. A. (1995) Chapter 9: Antiviral activity of cytokines. pp. 129-141. [0288]
  • Li, C. H. (1982) Mol. Cell. Biochem. 46: 31-41. [0289]
  • Lin, F.-K. (1996) U.S. Pat. No. 5,547,933. [0290]
  • Lin, F.-K., Suggs, S., Lin, C.-H., et al., Proc. Natl. Acad. Sci. USA (1985) 82: 7580-7584. [0291]
  • Linsley, P. S., Kallestad, J., Ochs, V. and Neubauer, M. (1990) 10: 1882-1890. [0292]
  • Livnah, O., Stura, E. A., Johnson, D. L. et al., (1996) Science 273: 464471. [0293]
  • Lu, H. S., Clogston, C. L., Narhi, L. O., Merewether, L. A., Pearl, W. R. and Boone, T. C. (1992) J. Biol. Chem. 267: 8770-8777. [0294]
  • Lydon, N. B., Favre, C., Bore, S., Neyret, O., Benureau, S., Levine, A. M., Seelig, G. F., Nagabhushan, T. L. and Trotta, P. P. (1985) Biochemistry 24: 4131-41. [0295]
  • MacGillivray, M. H., Baptista, J. and Johnson, A. (1996) J. Clin. Endocrinol. Metab. 81: 1806-1809. [0296]
  • Malik, N., Kallestad, J. C., Gunderson, N. L., Austin, S. D., Neubauer, M. G., Ochs, V., Marquardt, H., Zarling, J. M., Shoyab, M., Wei, C.-M., Linsley, P. S. and Rose, T. M. (1989) Molecular and Cellular Biology 9: 2847-2853. [0297]
  • Martial, J., Chene, N. and de la Llosa, P. (1985) FEBS. Letts. 180: 295-299. [0298]
  • Martial, J. A., Hallewell, R. A., Baxter, J. D. and Goodman, H. M. (1979) Science 205: 602-606. [0299]
  • Matthews, D. J., Topping, R. S., Cass, R. T. and Giebel, L. B. (1996) Proc. Natl. Acad. Sci. USA 93: 9471-9476. [0300]
  • McKay, D. B. (1992) Science 257: 412. [0301]
  • McKenzie, A. N. J., Culpepper, J. A., Malefyt, R. de W., Briere, F., Punnonen, J., Aversa, G., Sato, A., Dang, W., Cocks, B. G., Menon, S., de Vries, J. E., Banchereau, J., and Zurawski, G. (1993) Proc. Natl. Acad. Sci. USA 90: 3735-3739. [0302]
  • Meyers, F. J., Paradise, C., Scudder, S. A., Goodman, G. and Konrad, M. (1991) Clin. Pharmacol. Ther. 49: 307-313. [0303]
  • Milburn, M. V., Hassell, A. M., Lambert, M. H., Jordan, S. R., Proudfoot, A. E., Graber, P. and Wells, T. N. C. (1993) Nature 363: 172-176. [0304]
  • Mills, J. B., Kostyo, J. L., Reagan, C. R., Wagner, S. A., Moseley, M. H. and Wilhelm, A. E. (1980) Endocrinology 107: 391-399. [0305]
  • Minty, A., Chalon, P., Derocq, J.-M., Dumont, X., Guilemot, J.-C., Kaghad, M., Labit, C., Leplatois, P., Liauzun, P., Miloux, B., Minty, C., Casellas, P., Loison. G., Lupker, J., Shire, D., Ferrara, P. and Caput, D. (1993) Nature 362: 248-250. [0306]
  • Moreau, J.-F., Donaldson, D. D., Bennett, F., Witek-Giannotti, J., Clark, S. C. and Wong, G. G. (1988) Nature 336: 690-692. [0307]
  • Mott, H. R. and Campbell, I. D. (1995) Current Opinion in Structural Biology 5: 114-121. [0308]
  • Martial, J. A., Hallewell, R. A., Baxter, J. D. and Goodman, H. M. (1979) Science 205: 602-606. [0309]
  • Nagata, S. (1994) in Cytokines and Their Receptors, N. A. Nicola ed., Oxford University Press, Oxford, pp. 158-160. [0310]
  • Nagata, S., Tsuchiya, M., Asano, S., Kziro, Y., Yamazaki, T., Yamamoto, O., Hirata, Y., Kubota, N., Oh-eda, M., Nomura, H. and Ono, M. (1986a) Nature 319: 415-418. [0311]
  • Nagata, S., Tsuchiya, M., Asano, S., Yamamoto, O., Hirata, Y., Kubota, N., Onh-eda, M., Nomura, H. and Yamazaki, T. (1986b) EMBO J. 5: 575-581. [0312]
  • O'Reilly, D. R., Miller, L. K., Luckow, V. A. (1992) Caculovirus Expression Vectors, W. H. [0313]
  • Freeman Co. publishers, New York. [0314]
  • Otsuka, T., Miyajima, A., Brown, N., Otsu, K., Abrams, J., Saeland, S., Caux, C., De Waal-Malefyt, De Vries, J., Meyerson, P., Yokota, K., Gemmell, Rennick, D., Lee, F., Arai, N., Arai, K.-I. and Yokota, T. (1988) J. Immunology 140: 2288-2295. [0315]
  • Owers-Narhi, L., Arakawa, T., Aoki, K. H., Elmore, R., Rohde, M. F., Boone, T., Strickland, T. W. (1991) J. Biol. Chem. 266: 23022-23026. [0316]
  • Ozes, O. S., Reiter, Z., Klein, S., Blatt, L. M. and Taylor, M. W. (1992) J. Interferon Research 12: 55-59. [0317]
  • Paonessa, G., Graziani, R., de Serio, A., Savino, R., Ciapponi, L., Lahm, A., Ssalvati, A. L., Toniatti, C. and Ciliberto, G. (1995) EMBO J. 14: 1942-1951. [0318]
  • Park, L. S., Waldron, P. E., Friend, D., Sassenfeld, H. M., Price, V., Anderson, D., Cosman, D., Andrews, R. G., Bernstein, I. D. and Urdal, D. L. (1989) Blood 74: 56-65. [0319]
  • Paul, S. R., Bennett, F., Calvetti, J. A., Kelleher, K., Wood, C. R., O'Hara, R. M., Leary, A .C., Sibley, B., Clark, S. C., William, D. A. and Yang, Y.-C. (1990) Proc. Natl. Acad. Sci. USA 87: 7512-7516. [0320]
  • Pestka, S., Langer, J. A., Zoon, K. C. and Samuel, C. E. (1987) Ann. Rev. Biochem. 56: 727-777. [0321]
  • Pickering, L. A., Kronenberg, L. H. and Stewart, W. E., II (1980) Proc. Natl. Acad. Scio. USA 77: 5938-5942. [0322]
  • Powers, R., Garrett, D. S., March, C. J., Frieden, E. A., Gronenborn, A. M. and Clore, G. M. (1992) Science 256:1673-1677. [0323]
  • Radhakrishnan, R., Walter, L. J., Hruka, A., Reichert, P., Trotta, P. P., Nagabhushan, T. L. and Walter, M. R. (1996) Structure 4: 1453-1463. [0324]
  • Read, L. C., Tomas, F. M., Howarth, G. S., Martin, A. A., Edson, K. J., Gillespie, C. M., Owens, P. C. and Ballard, F. J. (1992) J. Endocrinology 133: 421431. [0325]
  • Redfield, C., Smith L. J., Boyd, J., Lawrence, G. M. P., Edwards, R. G., Smith, R. A. G., and Dobson, C. M. (1991) Biochemistry 30: 11029-11035. [0326]
  • Robinson, R. C., Grey, L. M., Stauton, D., Vankelecom, H., Vemallis, A. B., Moreau, J.-F., Stuart, D. I., Heath, J. K. and Jones, E. Y. (1994) Cell 77: 1101-1116. [0327]
  • Sasaki, H., Bothner, B., Dell, A. and Fukuda, M. (1987) J. Biol. Chem. 262 12059-12076. [0328]
  • Sasaki, H. Ochi, N., Dell, A. and Fukuda, M. (1988) Biochemistry 27: 8616-8626. [0329]
  • Shaw, G., Veldman, G. and Wooters, J. L. (1992) U.S. Pat. No. 5,166,322. [0330]
  • Shirafuji, N., Asano, S., Matsuda, S., Watari, K., Takaku, F. and Nagata, S. (1989) Exp Hematol. 17: 116-119. [0331]
  • Silvennoinen, O. and Ihle, J. N. (1996) Signalling by the Hematopoietic Cytokine Receptors, R. G. Landes, Company, Austin, Tex. [0332]
  • Souza, L. M., Boone, T. C., Gabrilove, J., Lai, P. H., Zsebo, K. M., Murdock, D. C., Chazin, V. R., Bruszewski, J., Lu, H., Chen, K. K., Barendt, J., Platzer, E., Moore, M. A. S., Mertelsmann, R. and Welte, K. (1986) Science 232: 61-65. [0333]
  • Spivak, J. L., Hogans, B. B. (1989) Blood 73: 90-99. [0334]
  • Takeuchi, M., Takasaki, S., Miyazaki, H., Takashi, D., Hoshi, S., Kochibe, N. and Kotaba, A. (1988) J. Biol. Chem. 263: 3657-3663. [0335]
  • Tanaka, H., Satake-Ishikawa, R., Ishikawa, M., Matsuki, S. and Asano, K. (1991) Cancer Research 51: 3710-3714. [0336]
  • Taniguchi, T., Matsui, H., Fujita, T., Takaoka, C., Kashima, N., Yoshimoto, R. and Hamuro, J. (1983) Nature 302: 305-310. [0337]
  • Taniguchi, T., Ohno, S., Fujii-Kuriyama, Y. and Muramatsu, M. (1980) Gene 10: 11-15. [0338]
  • Tamowski, S. J., Roy, S. K., Liptak, R. A., Lee, D. K. and Ning, R. Y. (1986) Methods Enzymology 119:153-165. [0339]
  • Tavernier, J., Tuypens, T., Verhee, A., Plaefinck, G., Devos, R., Van Der Heyden, J., Guisez, Y. and Oeffier, C. (1995) Proc. Natl. Acad. Sci. USA 92: 5194-5198. [0340]
  • Teh, L.-C. and Chapman, G. E. (1988) Biochem. Biophys. Res. Comm. 150: 391-398. [0341]
  • Thatcher, D. R. and Panayotatos, N. (1986) Methods Enzymology 119: 166-177. [0342]
  • Tomas, F. M., Knowles, S. E., Owens, P. C., Chandler, C. S., Francis, G. L., Read, L. C. and Ballard, F. J. (1992) Biochem. J. 282: 91-97. [0343]
  • Tsuchiya, M., Asano, S., Kaziro, Y and Nagata, S. (1986) Proc. Natl. Acad. Sci. USA 83: 7633-7637. [0344]
  • Tsuda, E., Kawanishi, G., Ueda, M., Masuda, S. and Sasaki, R. (1990) Eur. J. Biochem. 188: 405-411. [0345]
  • Vieira, P., de Waal-Malefyt, R., Dang, M. N., Johnson, K. E., Kastelein, R., Fiorentino, D. F., de Vries, J. E., Roncarolo, M.-G., Mosmann, T. R. and Moore, K. W. (1991) Proc. Natl. Acad. Sci. USA 88: 1172-1176. [0346]
  • Wang, A., Lu, S.-D., and Mark, D. F. (1984) Science 224: 1431-1433. [0347]
  • Walter, M. R., Cook, W. J., Ealick, S. E., Nagabhusan, T. L., Trotta, P. T. and Bugg, C. E. (1992) J. Mol. Biol. 224: 1075-1085. [0348]
  • Wen, D., Boissel, J.-P. R., Tracy, T. E., Gruninger, R. H., Mulcahy, L. S., Czelusniak, J., Goodman, M. and Bunn, H. F. (1993) Blood: 1507-1516. [0349]
  • Wen, D., Boissel, J. P., Showers, M., Ruch, B. C. and Bunn, H. F. (1994) J. Biol. Chem. 269: 22839-22846. [0350]
  • White, B. A., (1993), Methods in Molecular Biology, volume 15: PCR Protocols: Current Methods and Applications. Humana Press, Totowa, N.J. [0351]
  • Wojchowski, D. M., Orkin, S. H., Sytkowshi, A. J. (1987) Biochim. Biophys. Acta 910: 224-232. [0352]
  • Wolf, S. F., Temple, P. A., Kobayashi, M., Young, D., Dicig, M., Lowe, L., Dzialo, R., Fitz, L., Ferenz, C., Hewick, R. M., Kelleher, K., Herrmann, S. H., Clark, S. C., Azzoni, L., Chan, S. H., Trinchieri, G. and Perussia, B. (1991) J. Immunology 146: 3074-3081. [0353]
  • Wong, G. G. et al., (1987) Science 235: 1504. [0354]
  • Wrighton, N. C., Farrell, F. X., Chang, R. et al., (1996) Science 273: 458-463. [0355]
  • Yamaguchi, K., Akai, K., Kawanishi, G. Ueda, M., Masuda, S., Sasaki, R. (1991). J. Biol. Chem. 266: 20434-20439. [0356]
  • Yang, Y.-C., Ciarletta, A. B., Temple, P. A., Chung, M. P., Kovacic, S., Witek-Giannotti, J. S., Leary, A. C., Kriz, R., Donahue, R. E., Wong, G. G. and Clark, S. C. (1986) Cell 47: 3-10. [0357]
  • Yang, Y.-C., Ricciadi, S., Ciarletta, A., Calvetti, J., Kelleher, K. and Clark, S. C. (1989) Blood 74: 1880-1884. [0358]
  • Yokota, T., Otsuka, T., Mosmann, T., Banchereau, J., DeFrance, T., Blanchard, D., De Vries, J. E., Lee F. and Arai, K.-I. (1986) Proc. Natl. Acad. Sci. USA 83: 5894-5898. [0359]
  • Yokota, T., Coffinan, R. L., Hagiwara, H., Rennick, D. M., Takebe, Y., Yokota, K., Gemmell, L., Shrader, B., Yang, G., Meyerson, P., Luh, J., Hoy, P., Pene, J., Briere, F., Spits, H., Banchereau, J., De Vries, J., Lee, F., Arai, N. and Arai, K.-I. (1987) Proc. Natl. Acad. Sci. USA 84: 7388-7392. [0360]
  • Zurawski, S. M., Imler, J. L., and Zurawski, G. (1990) EMBO J. 9: 3899-3905. [0361]
  • Zurawski, S. M. and Zurawski, G. (1992) EMBO J. 11: 3905-3910. [0362]
  • 1 41 1 191 PRT Homo sapiens 1 Phe Pro Thr Ile Pro Leu Ser Arg Leu Phe Asp Asn Ala Met Leu Arg 1 5 10 15 Ala His Arg Leu His Gln Leu Ala Phe Asp Thr Tyr Gln Glu Phe Glu 20 25 30 Glu Ala Tyr Ile Pro Lys Glu Gln Lys Tyr Ser Phe Leu Gln Asn Pro 35 40 45 Gln Thr Ser Leu Cys Phe Ser Glu Ser Ile Pro Thr Pro Ser Asn Arg 50 55 60 Glu Glu Thr Gln Gln Lys Ser Asn Leu Glu Leu Leu Arg Ile Ser Leu 65 70 75 80 Leu Leu Ile Gln Ser Trp Leu Glu Pro Val Gln Phe Leu Arg Ser Val 85 90 95 Phe Ala Asn Ser Leu Val Tyr Gly Ala Ser Asp Ser Asn Val Tyr Asp 100 105 110 Leu Leu Lys Asp Leu Glu Glu Gly Ile Gln Thr Leu Met Gly Arg Leu 115 120 125 Glu Asp Gly Ser Pro Arg Thr Gly Gln Ile Phe Lys Gln Thr Tyr Ser 130 135 140 Lys Phe Asp Thr Asn Ser His Asn Asp Asp Ala Leu Leu Lys Asn Tyr 145 150 155 160 Gly Leu Leu Tyr Cys Phe Arg Lys Asp Met Asp Lys Val Glu Thr Phe 165 170 175 Leu Arg Ile Val Gln Cys Arg Ser Val Glu Gly Ser Cys Gly Phe 180 185 190 2 166 PRT Homo sapiens 2 Ala Pro Pro Arg Leu Ile Cys Asp Ser Arg Val Leu Glu Arg Tyr Leu 1 5 10 15 Leu Glu Ala Lys Glu Ala Glu Asn Ile Thr Thr Gly Cys Ala Glu His 20 25 30 Cys Ser Leu Asn Glu Asn Ile Thr Val Pro Asp Thr Lys Val Asn Phe 35 40 45 Tyr Ala Trp Lys Arg Met Glu Val Gly Gln Gln Ala Val Glu Val Trp 50 55 60 Gln Gly Leu Ala Leu Leu Ser Glu Ala Val Leu Arg Gly Gln Ala Leu 65 70 75 80 Leu Val Asn Ser Ser Gln Pro Trp Glu Pro Leu Gln Leu His Val Asp 85 90 95 Lys Ala Val Ser Gly Leu Arg Ser Leu Thr Thr Leu Leu Arg Ala Leu 100 105 110 Gly Ala Gln Lys Glu Ala Ile Ser Pro Pro Asp Ala Ala Ser Ala Ala 115 120 125 Pro Leu Arg Thr Ile Thr Ala Asp Thr Phe Arg Lys Leu Phe Arg Val 130 135 140 Tyr Ser Asn Phe Leu Arg Gly Lys Leu Lys Leu Tyr Thr Gly Glu Ala 145 150 155 160 Cys Arg Thr Gly Asp Arg 165 3 165 PRT Homo sapiens 3 Cys Asp Leu Pro Gln Thr His Ser Leu Gly Ser Arg Arg Thr Leu Met 1 5 10 15 Leu Leu Ala Gln Met Arg Arg Ile Ser Leu Phe Ser Cys Leu Lys Asp 20 25 30 Arg His Asp Phe Gly Phe Pro Gln Glu Glu Phe Gly Asn Gln Phe Gln 35 40 45 Lys Ala Glu Thr Ile Pro Val Leu His Glu Met Ile Gln Gln Ile Phe 50 55 60 Asn Leu Phe Ser Thr Lys Asp Ser Ser Ala Ala Trp Asp Glu Thr Leu 65 70 75 80 Leu Asp Lys Phe Tyr Thr Glu Leu Tyr Gln Gln Leu Asn Asp Leu Glu 85 90 95 Ala Cys Val Ile Gln Gly Val Gly Val Thr Glu Thr Pro Leu Met Lys 100 105 110 Glu Asp Ser Ile Leu Ala Val Arg Lys Tyr Phe Gln Arg Ile Thr Leu 115 120 125 Tyr Leu Lys Glu Lys Lys Tyr Ser Pro Cys Ala Trp Glu Val Val Arg 130 135 140 Ala Glu Ile Met Arg Ser Phe Ser Leu Ser Thr Asn Leu Gln Glu Ser 145 150 155 160 Leu Arg Ser Lys Glu 165 4 166 PRT Homo sapiens 4 Cys Asp Leu Pro Glu Thr His Ser Leu Asp Asn Arg Arg Thr Leu Met 1 5 10 15 Leu Leu Ala Gln Met Ser Arg Ile Ser Pro Ser Ser Cys Leu Met Asp 20 25 30 Arg His Asp Phe Gly Phe Pro Gln Glu Glu Phe Asp Gly Asn Gln Phe 35 40 45 Gln Lys Ala Pro Ala Ile Ser Val Leu His Glu Leu Ile Gln Gln Ile 50 55 60 Phe Asn Leu Phe Thr Thr Lys Asp Ser Ser Ala Ala Trp Asp Glu Asp 65 70 75 80 Leu Leu Asp Lys Phe Cys Thr Glu Leu Tyr Gln Gln Leu Asn Asp Leu 85 90 95 Glu Ala Cys Val Met Gln Glu Glu Arg Val Gly Glu Thr Pro Leu Met 100 105 110 Asn Ala Asp Ser Ile Leu Ala Val Lys Lys Tyr Phe Arg Arg Ile Thr 115 120 125 Leu Tyr Leu Thr Glu Lys Lys Tyr Ser Pro Cys Ala Trp Glu Val Val 130 135 140 Arg Ala Glu Ile Met Arg Ser Leu Ser Leu Ser Thr Asn Leu Gln Glu 145 150 155 160 Arg Leu Arg Arg Lys Glu 165 5 166 PRT Homo sapiens 5 Met Ser Tyr Asn Leu Leu Gly Phe Leu Gln Arg Ser Ser Asn Phe Gln 1 5 10 15 Cys Gln Lys Leu Leu Trp Gln Leu Asn Gly Arg Leu Glu Tyr Cys Leu 20 25 30 Lys Asp Arg Met Asn Phe Asp Ile Pro Glu Glu Ile Lys Gln Leu Gln 35 40 45 Gln Phe Gln Lys Glu Asp Ala Ala Leu Thr Ile Tyr Glu Met Leu Gln 50 55 60 Asn Ile Phe Ala Ile Phe Arg Gln Asp Ser Ser Ser Thr Gly Trp Asn 65 70 75 80 Glu Thr Ile Val Glu Asn Leu Leu Ala Asn Val Tyr His Gln Ile Asn 85 90 95 His Leu Lys Thr Val Leu Glu Glu Lys Leu Glu Lys Glu Asp Phe Thr 100 105 110 Arg Gly Lys Leu Met Ser Ser Leu His Leu Lys Arg Tyr Tyr Gly Arg 115 120 125 Ile Leu His Tyr Leu Lys Ala Lys Glu Tyr Ser His Cys Ala Trp Thr 130 135 140 Ile Val Arg Val Glu Ile Leu Arg Asn Phe Tyr Phe Ile Asn Arg Leu 145 150 155 160 Thr Gly Tyr Leu Arg Asn 165 6 174 PRT Homo sapiens 6 Thr Pro Leu Gly Pro Ala Ser Ser Leu Pro Gln Ser Phe Leu Leu Lys 1 5 10 15 Cys Leu Glu Gln Val Arg Lys Ile Gln Gly Asp Gly Ala Ala Leu Gln 20 25 30 Glu Lys Leu Cys Ala Thr Tyr Lys Leu Cys His Pro Glu Glu Leu Val 35 40 45 Leu Leu Gly His Ser Leu Gly Ile Pro Trp Ala Pro Leu Ser Ser Cys 50 55 60 Pro Ser Gln Ala Leu Gln Leu Ala Gly Cys Leu Ser Gln Leu His Ser 65 70 75 80 Gly Leu Phe Leu Tyr Gln Gly Leu Leu Gln Ala Leu Glu Gly Ile Ser 85 90 95 Pro Glu Leu Gly Pro Thr Leu Asp Thr Leu Gln Leu Asp Val Ala Asp 100 105 110 Phe Ala Thr Thr Ile Trp Gln Gln Met Glu Glu Leu Gly Met Ala Pro 115 120 125 Ala Leu Gln Pro Thr Gln Gly Ala Met Pro Ala Phe Ala Ser Ala Phe 130 135 140 Gln Arg Arg Ala Gly Gly Val Leu Val Ala Ser His Leu Gln Ser Phe 145 150 155 160 Leu Glu Val Ser Tyr Arg Val Leu Arg His Leu Ala Gln Pro 165 170 7 332 PRT Homo sapiens 7 Ser Pro Ala Pro Pro Ala Cys Asp Leu Arg Val Leu Ser Lys Leu Leu 1 5 10 15 Arg Asp Ser His Val Leu His Ser Arg Leu Ser Gln Cys Pro Glu Val 20 25 30 His Pro Leu Pro Thr Pro Val Leu Leu Pro Ala Val Asp Phe Ser Leu 35 40 45 Gly Glu Trp Lys Thr Gln Met Glu Glu Thr Lys Ala Gln Asp Ile Leu 50 55 60 Gly Ala Val Thr Leu Leu Leu Glu Gly Val Met Ala Ala Arg Gly Gln 65 70 75 80 Leu Gly Pro Thr Cys Leu Ser Ser Leu Leu Gly Gln Leu Ser Gly Gln 85 90 95 Val Arg Leu Leu Leu Gly Ala Leu Gln Ser Leu Leu Gly Thr Gln Leu 100 105 110 Pro Pro Gln Gly Arg Thr Thr Ala His Lys Asp Pro Asn Ala Ile Phe 115 120 125 Leu Ser Phe Gln His Leu Leu Arg Gly Lys Val Arg Phe Leu Met Leu 130 135 140 Val Gly Gly Ser Thr Leu Cys Val Arg Arg Ala Pro Pro Thr Thr Ala 145 150 155 160 Val Pro Ser Arg Thr Ser Leu Val Leu Thr Leu Asn Glu Leu Pro Asn 165 170 175 Arg Thr Ser Gly Leu Leu Glu Thr Asn Phe Thr Ala Ser Ala Arg Thr 180 185 190 Thr Gly Ser Gly Leu Leu Lys Trp Gln Gln Gly Phe Arg Ala Lys Ile 195 200 205 Pro Gly Leu Leu Asn Gln Thr Ser Arg Ser Leu Asp Gln Ile Pro Gly 210 215 220 Tyr Leu Asn Arg Ile His Glu Leu Leu Asn Gly Thr Arg Gly Leu Phe 225 230 235 240 Pro Gly Pro Ser Arg Arg Thr Leu Gly Ala Pro Asp Ile Ser Ser Gly 245 250 255 Thr Ser Asp Thr Gly Ser Leu Pro Pro Asn Leu Gln Pro Gly Tyr Ser 260 265 270 Pro Ser Pro Thr His Pro Pro Thr Gly Gly Tyr Thr Leu Phe Pro Leu 275 280 285 Pro Pro Thr Leu Pro Thr Pro Val Val Gln Leu His Pro Leu Leu Pro 290 295 300 Asp Pro Ser Ala Pro Thr Pro Thr Pro Thr Ser Pro Leu Leu Asn Thr 305 310 315 320 Ser Tyr Thr His Ser Gln Asn Leu Ser Gln Glu Gly 325 330 8 127 PRT Homo sapiens 8 Ala Pro Ala Arg Ser Pro Ser Pro Ser Thr Gln Pro Trp Glu His Val 1 5 10 15 Asn Ala Ile Gln Glu Ala Arg Arg Leu Leu Asn Leu Ser Arg Asp Thr 20 25 30 Ala Ala Glu Met Asn Glu Thr Val Glu Val Ile Ser Glu Met Phe Asp 35 40 45 Leu Gln Glu Pro Thr Cys Leu Gln Thr Arg Leu Glu Leu Tyr Lys Gln 50 55 60 Gly Leu Arg Gly Ser Leu Thr Lys Leu Lys Gly Pro Leu Thr Met Met 65 70 75 80 Ala Ser His Tyr Lys Gln His Cys Pro Pro Thr Pro Glu Thr Ser Cys 85 90 95 Ala Thr Gln Ile Ile Thr Phe Glu Ser Phe Lys Glu Asn Leu Lys Asp 100 105 110 Phe Leu Leu Val Ile Pro Phe Asp Cys Trp Glu Pro Val Gln Glu 115 120 125 9 133 PRT Homo sapiens 9 Ala Pro Thr Ser Ser Ser Thr Lys Lys Thr Gln Leu Gln Leu Glu His 1 5 10 15 Leu Leu Leu Asp Leu Gln Met Ile Leu Asn Gly Ile Asn Asn Tyr Lys 20 25 30 Asn Pro Lys Leu Thr Arg Met Leu Thr Phe Lys Phe Tyr Met Pro Lys 35 40 45 Lys Ala Thr Glu Leu Lys His Leu Gln Cys Leu Glu Glu Glu Leu Lys 50 55 60 Pro Leu Glu Glu Val Leu Asn Leu Ala Gln Ser Lys Asn Phe His Leu 65 70 75 80 Arg Pro Arg Asp Leu Ile Ser Asn Ile Asn Val Ile Val Leu Glu Leu 85 90 95 Lys Gly Ser Glu Thr Thr Phe Met Cys Glu Tyr Ala Asp Glu Thr Ala 100 105 110 Thr Ile Val Glu Phe Leu Asn Arg Trp Ile Thr Phe Cys Gln Ser Ile 115 120 125 Ile Ser Thr Leu Thr 130 10 152 PRT Homo sapiens 10 Met Ser Arg Leu Pro Val Leu Leu Leu Leu Gln Leu Leu Val Arg Pro 1 5 10 15 Gly Leu Gln Ala Pro Met Thr Gln Thr Thr Pro Leu Lys Thr Ser Trp 20 25 30 Val Asn Cys Ser Asn Met Ile Asp Glu Ile Ile Thr His Leu Lys Gln 35 40 45 Pro Pro Leu Pro Leu Leu Asp Phe Asn Asn Leu Asn Gly Glu Asp Gln 50 55 60 Asp Ile Leu Met Glu Asn Asn Leu Arg Arg Pro Asn Leu Glu Ala Phe 65 70 75 80 Asn Arg Ala Val Lys Ser Leu Gln Asn Ala Ser Ala Ile Glu Ser Ile 85 90 95 Leu Lys Asn Leu Leu Pro Cys Leu Pro Leu Ala Thr Ala Ala Pro Thr 100 105 110 Arg His Pro Ile His Ile Lys Asp Gly Asp Trp Asn Glu Phe Arg Arg 115 120 125 Lys Leu Thr Phe Tyr Leu Lys Thr Leu Glu Asn Ala Gln Ala Gln Gln 130 135 140 Thr Thr Leu Ser Leu Ala Ile Phe 145 150 11 129 PRT Homo sapiens 11 His Lys Cys Asp Ile Thr Leu Gln Glu Ile Ile Lys Thr Leu Asn Ser 1 5 10 15 Leu Thr Glu Gln Lys Thr Leu Cys Thr Glu Leu Thr Val Thr Asp Ile 20 25 30 Phe Ala Ala Ser Lys Asn Thr Thr Glu Lys Glu Thr Phe Cys Arg Ala 35 40 45 Ala Thr Val Leu Arg Gln Phe Tyr Ser His His Glu Lys Asp Thr Arg 50 55 60 Cys Leu Gly Ala Thr Ala Gln Gln Phe His Arg His Lys Gln Leu Ile 65 70 75 80 Arg Phe Leu Lys Arg Leu Asp Arg Asn Leu Trp Gly Leu Ala Gly Leu 85 90 95 Asn Ser Cys Pro Val Lys Glu Ala Asn Gln Ser Thr Leu Glu Asn Phe 100 105 110 Leu Glu Arg Leu Lys Thr Ile Met Arg Glu Lys Tyr Ser Lys Cys Ser 115 120 125 Ser 12 134 PRT Homo sapiens 12 Met Arg Met Leu Leu His Leu Ser Leu Leu Ala Leu Gly Ala Ala Tyr 1 5 10 15 Val Tyr Ala Ile Pro Thr Glu Ile Pro Thr Ser Ala Leu Val Lys Glu 20 25 30 Thr Leu Ala Leu Leu Ser Thr His Arg Thr Leu Leu Ile Ala Asn Glu 35 40 45 Thr Leu Arg Ile Pro Val Pro Val His Lys Asn His Gln Leu Cys Thr 50 55 60 Glu Glu Ile Phe Gln Gly Ile Gly Thr Leu Glu Ser Gln Thr Val Gln 65 70 75 80 Gly Gly Thr Val Glu Arg Leu Phe Lys Asn Leu Ser Leu Ile Lys Lys 85 90 95 Tyr Ile Asp Gly Gln Lys Lys Lys Cys Gly Glu Glu Arg Arg Arg Val 100 105 110 Asn Gln Phe Leu Asp Tyr Leu Gln Glu Phe Leu Gly Val Met Asn Thr 115 120 125 Glu Trp Ile Ile Glu Ser 130 13 212 PRT Homo sapiens 13 Met Asn Ser Phe Ser Thr Ser Ala Phe Gly Pro Val Ala Phe Ser Leu 1 5 10 15 Gly Leu Leu Leu Val Leu Pro Ala Ala Phe Pro Ala Pro Val Pro Pro 20 25 30 Gly Glu Asp Ser Lys Asp Val Ala Ala Pro His Arg Gln Pro Leu Thr 35 40 45 Ser Ser Glu Arg Ile Asp Lys Gln Ile Arg Tyr Ile Leu Asp Gly Ile 50 55 60 Ser Ala Leu Arg Lys Glu Thr Cys Asn Lys Ser Asn Met Cys Glu Ser 65 70 75 80 Ser Lys Glu Ala Leu Ala Glu Asn Asn Leu Asn Leu Pro Lys Met Ala 85 90 95 Glu Lys Asp Gly Cys Phe Gln Ser Gly Phe Asn Glu Glu Thr Cys Leu 100 105 110 Val Lys Ile Ile Thr Gly Leu Leu Glu Phe Glu Val Tyr Leu Glu Tyr 115 120 125 Leu Gln Asn Arg Phe Glu Ser Ser Glu Glu Gln Ala Arg Ala Val Gln 130 135 140 Met Ser Thr Lys Val Leu Ile Gln Phe Leu Gln Lys Lys Ala Lys Asn 145 150 155 160 Leu Asp Ala Ile Thr Thr Pro Asp Pro Thr Thr Asn Ala Ser Leu Leu 165 170 175 Thr Lys Leu Gln Ala Gln Asn Gln Trp Leu Gln Asp Met Thr Thr His 180 185 190 Leu Ile Leu Arg Ser Phe Lys Glu Phe Leu Gln Ser Ser Leu Arg Ala 195 200 205 Leu Arg Gln Met 210 14 177 PRT Homo sapiens 14 Met Phe His Val Ser Phe Arg Tyr Ile Phe Gly Leu Pro Pro Leu Ile 1 5 10 15 Leu Val Leu Leu Pro Val Ala Ser Ser Asp Cys Asp Ile Glu Gly Lys 20 25 30 Asp Gly Lys Gln Tyr Glu Ser Val Leu Met Val Ser Ile Asp Gln Leu 35 40 45 Leu Asp Ser Met Lys Glu Ile Gly Ser Asn Cys Leu Asn Asn Glu Phe 50 55 60 Asn Phe Phe Lys Arg His Ile Cys Asp Ala Asn Lys Glu Gly Met Phe 65 70 75 80 Leu Phe Arg Ala Ala Arg Lys Leu Arg Gln Phe Leu Lys Met Asn Ser 85 90 95 Thr Gly Asp Phe Asp Leu His Leu Leu Lys Val Ser Glu Gly Thr Thr 100 105 110 Ile Leu Leu Asn Cys Thr Gly Gln Val Lys Gly Arg Lys Pro Ala Ala 115 120 125 Leu Gly Glu Ala Gln Pro Thr Lys Ser Leu Glu Glu Asn Lys Ser Leu 130 135 140 Lys Glu Gln Lys Lys Leu Asn Asp Leu Cys Phe Leu Lys Arg Leu Leu 145 150 155 160 Gln Glu Ile Lys Thr Cys Trp Asn Lys Ile Leu Met Gly Thr Lys Glu 165 170 175 His 15 144 PRT Homo sapiens 15 Met Leu Leu Ala Met Val Leu Thr Ser Ala Leu Leu Leu Cys Ser Val 1 5 10 15 Ala Gly Gln Gly Cys Pro Thr Leu Ala Gly Ile Leu Asp Ile Asn Phe 20 25 30 Leu Ile Asn Lys Met Gln Glu Asp Pro Ala Ser Lys Cys His Cys Ser 35 40 45 Ala Asn Val Thr Ser Cys Leu Cys Leu Gly Ile Pro Ser Asp Asn Cys 50 55 60 Thr Arg Pro Cys Phe Ser Glu Arg Leu Ser Gln Met Thr Asn Thr Thr 65 70 75 80 Met Gln Thr Arg Tyr Pro Leu Ile Phe Ser Arg Val Lys Lys Ser Val 85 90 95 Glu Val Leu Lys Asn Asn Lys Cys Pro Tyr Phe Ser Cys Glu Gln Pro 100 105 110 Cys Asn Gln Thr Thr Ala Gly Asn Ala Leu Thr Phe Leu Lys Ser Leu 115 120 125 Leu Glu Ile Phe Gln Lys Glu Lys Met Arg Gly Met Arg Gly Lys Ile 130 135 140 16 178 PRT Homo sapiens 16 Met His Ser Ser Ala Leu Leu Cys Cys Leu Val Leu Leu Thr Gly Val 1 5 10 15 Arg Ala Ser Pro Gly Gln Gly Thr Gln Ser Glu Asn Ser Cys Thr His 20 25 30 Phe Pro Gly Asn Leu Pro Asn Met Leu Arg Asp Leu Arg Asp Ala Phe 35 40 45 Ser Arg Val Lys Thr Phe Phe Gln Met Lys Asp Gln Leu Asp Asn Leu 50 55 60 Leu Leu Lys Glu Ser Leu Leu Glu Asp Phe Lys Gly Tyr Leu Gly Cys 65 70 75 80 Gln Ala Leu Ser Glu Met Ile Gln Phe Tyr Leu Glu Glu Val Met Pro 85 90 95 Gln Ala Glu Asn Gln Asp Pro Asp Ile Lys Ala His Val Asn Ser Leu 100 105 110 Gly Glu Asn Leu Lys Thr Leu Arg Leu Arg Leu Arg Arg Cys His Arg 115 120 125 Phe Leu Pro Cys Glu Asn Lys Ser Lys Ala Val Glu Gln Val Lys Asn 130 135 140 Ala Phe Asn Lys Leu Gln Glu Lys Gly Ile Tyr Lys Ala Met Ser Glu 145 150 155 160 Phe Asp Ile Phe Ile Asn Tyr Ile Glu Ala Tyr Met Thr Met Lys Ile 165 170 175 Arg Asn 17 199 PRT Homo sapiens 17 Met Asn Cys Val Cys Arg Leu Val Leu Val Val Leu Ser Leu Trp Pro 1 5 10 15 Asp Thr Ala Val Ala Pro Gly Pro Pro Pro Gly Pro Pro Arg Val Ser 20 25 30 Pro Asp Pro Arg Ala Glu Leu Asp Ser Thr Val Leu Leu Thr Arg Ser 35 40 45 Leu Leu Ala Asp Thr Arg Gln Leu Ala Ala Gln Leu Arg Asp Lys Phe 50 55 60 Pro Ala Asp Gly Asp His Asn Leu Asp Ser Leu Pro Thr Leu Ala Met 65 70 75 80 Ser Ala Gly Ala Leu Gly Ala Leu Gln Leu Pro Gly Val Leu Thr Arg 85 90 95 Leu Arg Ala Asp Leu Leu Ser Tyr Leu Arg His Val Gln Trp Leu Arg 100 105 110 Arg Ala Gly Gly Ser Ser Leu Lys Thr Leu Glu Pro Glu Leu Gly Thr 115 120 125 Leu Gln Ala Arg Leu Asp Arg Leu Leu Arg Arg Leu Gln Leu Leu Met 130 135 140 Ser Arg Leu Ala Leu Pro Gln Pro Pro Pro Asp Pro Pro Ala Pro Pro 145 150 155 160 Leu Ala Pro Pro Ser Ser Ala Trp Gly Gly Ile Arg Ala Ala His Ala 165 170 175 Ile Leu Gly Gly Leu His Leu Thr Leu Asp Trp Ala Val Arg Gly Leu 180 185 190 Leu Leu Leu Lys Thr Arg Leu 195 18 219 PRT Homo sapiens 18 Met Cys Pro Ala Arg Ser Leu Leu Leu Val Ala Thr Leu Val Leu Leu 1 5 10 15 Asp His Leu Ser Leu Ala Arg Asn Leu Pro Val Ala Thr Pro Asp Pro 20 25 30 Gly Met Phe Pro Cys Leu His His Ser Gln Asn Leu Leu Arg Ala Val 35 40 45 Ser Asn Met Leu Gln Lys Ala Arg Gln Thr Leu Glu Phe Tyr Pro Cys 50 55 60 Thr Ser Glu Glu Ile Asp His Glu Asp Ile Thr Lys Asp Lys Thr Ser 65 70 75 80 Thr Val Glu Ala Cys Leu Pro Leu Glu Leu Thr Lys Asn Glu Ser Cys 85 90 95 Leu Asn Ser Arg Glu Thr Ser Phe Ile Thr Asn Gly Ser Cys Leu Ala 100 105 110 Ser Arg Lys Thr Ser Phe Met Met Ala Leu Cys Leu Ser Ser Ile Tyr 115 120 125 Glu Asp Leu Lys Met Tyr Gln Val Glu Phe Lys Thr Met Asn Ala Lys 130 135 140 Leu Leu Met Asp Pro Lys Arg Gln Ile Phe Leu Asp Gln Asn Met Leu 145 150 155 160 Ala Val Ile Asp Glu Leu Met Gln Ala Leu Asn Phe Asn Ser Glu Thr 165 170 175 Val Pro Gln Lys Ser Ser Leu Glu Glu Pro Asp Phe Tyr Lys Thr Lys 180 185 190 Ile Lys Leu Cys Ile Leu Leu His Ala Phe Arg Ile Arg Ala Val Thr 195 200 205 Ile Asp Arg Val Thr Ser Tyr Leu Asn Ala Ser 210 215 19 132 PRT Homo sapiens 19 Met Ala Leu Leu Leu Thr Thr Val Ile Ala Leu Thr Cys Leu Gly Gly 1 5 10 15 Phe Ala Ser Pro Gly Pro Val Pro Pro Ser Thr Ala Leu Arg Glu Leu 20 25 30 Ile Glu Glu Leu Val Asn Ile Thr Gln Asn Gln Lys Ala Pro Leu Cys 35 40 45 Asn Gly Ser Met Val Trp Ser Ile Asn Leu Thr Ala Gly Met Tyr Cys 50 55 60 Ala Ala Leu Glu Ser Leu Ile Asn Val Ser Gly Cys Ser Ala Ile Glu 65 70 75 80 Lys Thr Gln Arg Met Leu Ser Gly Phe Cys Pro His Lys Val Ser Ala 85 90 95 Gly Gln Phe Ser Ser Leu His Val Arg Asp Thr Lys Ile Glu Val Ala 100 105 110 Gln Phe Val Lys Asp Leu Leu Leu His Leu Lys Lys Leu Phe Arg Glu 115 120 125 Gly Arg Phe Asn 130 20 114 PRT Homo sapiens 20 Asn Trp Val Asn Val Ile Ser Asp Leu Lys Lys Ile Glu Asp Leu Ile 1 5 10 15 Gln Ser Met His Ile Asp Ala Thr Leu Tyr Thr Glu Ser Asp Val His 20 25 30 Pro Ser Cys Lys Val Thr Ala Met Lys Cys Phe Leu Leu Glu Leu Gln 35 40 45 Val Ile Ser Leu Glu Ser Gly Asp Ala Ser Ile His Asp Thr Val Glu 50 55 60 Asn Leu Ile Ile Leu Ala Asn Asn Ser Leu Ser Ser Asn Gly Asn Val 65 70 75 80 Thr Glu Ser Gly Cys Lys Glu Cys Glu Glu Leu Glu Glu Lys Asn Ile 85 90 95 Lys Glu Phe Leu Gln Ser Phe Val His Ile Val Gln Met Phe Ile Asn 100 105 110 Thr Ser 21 252 PRT Homo sapiens 21 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala 1 5 10 15 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Ala Ile Gly Ser Cys Ser 20 25 30 Lys Glu Tyr Arg Val Leu Leu Gly Gln Leu Gln Lys Gln Thr Asp Leu 35 40 45 Met Gln Asp Thr Ser Arg Leu Leu Asp Pro Tyr Ile Arg Ile Gln Gly 50 55 60 Leu Asp Val Pro Lys Leu Arg Glu His Cys Arg Glu Arg Pro Gly Ala 65 70 75 80 Phe Pro Ser Glu Glu Thr Leu Arg Gly Leu Gly Arg Arg Gly Phe Leu 85 90 95 Gln Thr Leu Asn Ala Thr Leu Gly Cys Val Leu His Arg Leu Ala Asp 100 105 110 Leu Glu Gln Arg Leu Pro Lys Ala Gln Asp Leu Glu Arg Ser Gly Leu 115 120 125 Asn Ile Glu Asp Leu Glu Lys Leu Gln Met Ala Arg Pro Asn Ile Leu 130 135 140 Gly Leu Arg Asn Asn Ile Tyr Cys Met Ala Gln Leu Leu Asp Asn Ser 145 150 155 160 Asp Thr Ala Glu Pro Thr Lys Ala Gly Arg Gly Ala Ser Gln Pro Pro 165 170 175 Thr Pro Thr Pro Ala Ser Asp Ala Phe Gln Arg Lys Leu Glu Gly Cys 180 185 190 Arg Phe Leu His Gly Tyr His Arg Phe Met His Ser Val Gly Arg Val 195 200 205 Phe Ser Lys Trp Gly Glu Ser Pro Asn Arg Ser Arg Arg His Ser Pro 210 215 220 His Gln Ala Leu Arg Lys Gly Val Arg Arg Thr Arg Pro Ser Arg Lys 225 230 235 240 Gly Lys Arg Leu Met Thr Arg Gly Gln Leu Pro Arg 245 250 22 200 PRT Homo sapiens 22 Met Ala Phe Thr Glu His Ser Pro Leu Thr Pro His Arg Arg Asp Leu 1 5 10 15 Cys Ser Arg Ser Ile Trp Leu Ala Arg Lys Ile Arg Ser Asp Leu Thr 20 25 30 Ala Leu Thr Glu Ser Tyr Val Lys His Gln Gly Leu Asn Lys Asn Ile 35 40 45 Asn Leu Asp Ser Ala Asp Gly Met Pro Val Ala Ser Thr Asp Gln Trp 50 55 60 Ser Glu Leu Thr Glu Ala Glu Arg Leu Gln Glu Asn Leu Gln Ala Tyr 65 70 75 80 Arg Thr Phe His Val Leu Leu Ala Arg Leu Leu Glu Asp Gln Gln Val 85 90 95 His Phe Thr Pro Thr Glu Gly Asp Phe His Gln Ala Ile His Thr Leu 100 105 110 Leu Leu Gln Val Ala Ala Phe Ala Tyr Gln Ile Glu Glu Leu Met Ile 115 120 125 Leu Leu Glu Tyr Lys Ile Pro Arg Asn Glu Ala Asp Gly Met Pro Ile 130 135 140 Asn Val Gly Asp Gly Gly Leu Phe Glu Lys Lys Leu Trp Gly Leu Lys 145 150 155 160 Val Leu Gln Glu Leu Ser Gln Trp Thr Val Arg Ser Ile His Asp Leu 165 170 175 Arg Phe Ile Ser Ser His Gln Thr Gly Ile Pro Ala Arg Gly Ser His 180 185 190 Tyr Ile Ala Asn Asn Lys Lys Met 195 200 23 181 PRT Homo sapiens 23 Ser Pro Leu Pro Ile Thr Pro Val Asn Ala Thr Cys Ala Ile Arg His 1 5 10 15 Pro Cys His Asn Asn Leu Met Asn Gln Ile Arg Ser Gln Leu Ala Gln 20 25 30 Leu Asn Gly Ser Ala Asn Ala Leu Phe Ile Leu Tyr Tyr Thr Ala Gln 35 40 45 Gly Glu Pro Phe Pro Asn Asn Leu Asp Lys Leu Cys Gly Pro Asn Val 50 55 60 Thr Asp Phe Pro Pro Phe His Ala Asn Gly Thr Glu Lys Ala Lys Leu 65 70 75 80 Val Glu Leu Tyr Arg Ile Val Val Tyr Leu Gly Thr Ser Leu Gly Asn 85 90 95 Ile Thr Arg Asp Gln Lys Ile Leu Asn Pro Ser Ala Leu Ser Leu His 100 105 110 Ser Lys Leu Asn Ala Thr Ala Asp Ile Leu Arg Gly Leu Leu Ser Asn 115 120 125 Val Leu Cys Arg Leu Cys Ser Lys Tyr His Val Gly His Val Asp Val 130 135 140 Thr Tyr Gly Pro Pro Asp Thr Ser Gly Lys Asp Val Phe Gln Lys Lys 145 150 155 160 Lys Leu Gly Cys Gln Leu Leu Gly Lys Tyr Lys Gln Ile Ile Ala Val 165 170 175 Leu Ala Gln Ala Phe 180 24 29 DNA Artificial Sequence Description of Artificial SequencePCR Primer 24 catatgttcc caaccattcc cttatccag 29 25 33 DNA Artificial Sequence Description of Artificial SequencePCR Primer 25 gggggatcct cactagaagc cacagctgcc ctc 33 26 39 DNA Artificial Sequence Description of Artificial SequencePCR Primer 26 ccccggatcc gccaccatgg atctctggca gctgctgtt 39 27 40 DNA Artificial Sequence Description of Artificial SequencePCR Primer 27 ccccgtcgac tctagagcta ttaaatacgt agctcttggg 40 28 32 DNA Artificial Sequence Description of Artificial SequencePCR Primer 28 cgcggatccg attagaatcc acagctcccc tc 32 29 66 DNA Artificial Sequence Description of Artificial SequencePCR Primer 29 ccccctctag acatatgaag aagaacatcg cattcctgct ggcatctatg ttcgttttct 60 ctatcg 66 30 65 DNA Artificial Sequence Description of Artificial SequencePCR Primer 30 gcatctatgt tcgttttctc tatcgctacc aacgcttacg cattcccaac cattccctta 60 tccag 65 31 62 DNA Artificial Sequence Description of Artificial SequencePCR Primer 31 gcagtggcac tggctggttt cgctaccgta gcgcaggcct tcccaaccat tcccttatcc 60 ag 62 32 59 DNA Artificial Sequence Description of Artificial SequencePCR Primer 32 ccccgtcgac acatatgaag aagacagcta tcgcgattgc agtggcactg gctggtttc 59 33 36 DNA Artificial Sequence Description of Artificial SequencePCR Primer 33 ctgcttgaag atctgcccac accgggggct gccatc 36 34 24 DNA Artificial Sequence Description of Artificial SequencePCR Primer 34 gtagcgcagg ccttcccaac catt 24 35 39 DNA Artificial Sequence Description of Artificial SequencePCR Primer 35 ctgcttgaag atctgcccag tccgggggca gccatcttc 39 36 51 DNA Artificial Sequence Description of Artificial SequencePCR Primer 36 gggcagatct tcaagcagac ctacagcaag ttcgactgca actcacacaa c 51 37 34 DNA Artificial Sequence Description of Artificial SequencePCR Primer 37 cgcggtaccc gggatccgat tagaatccac agct 34 38 36 DNA Artificial Sequence Description of Artificial SequencePCR Primer 38 gggcagatct tcaagcagac ctactgcaag ttcgac 36 39 42 DNA Artificial Sequence Description of Artificial SequencePCR Primer 39 cgcggtaccg gatccttagc agaagccaca gctgccctcc ac 42 40 24 DNA Artificial Sequence Description of Artificial SequencePCR Primer 40 gtagcgcagg ccttcccaac catt 24 41 40 DNA Artificial Sequence Description of Artificial SequencePCR Primer 41 ccccgtcgac tctagagcca ttagatacaa agctcttggg 40

Claims (59)

What is claimed is:
1. A cysteine variant of granulocyte colony-stimulating factor of SEQ ID NO:6, wherein a cysteine residue is substituted for at least one amino acid located in at least one region of granulocyte colony-stimulating factor selected from the group consisting of: the A-B loop, the B-C loop, the first three or last three amino acids in helix B, the first three or last three amino acids in helix C, the first three or last three amino acids in helix D, and the amino acids following helix D; wherein said variant has biological activity in vitro as measured by proliferation of a cell line that proliferates in response to granulocyte colony-stimulating factor.
2. The cysteine variant according to claim 1, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of: K40, S53, G55, W58, A59, P60, S62, S63, P65, S66, Q67, A68, Q70, A72, Q90, A91, E93, G94, S96, E98, G100, A143, Q145, Q173 and P174.
3. A cysteine variant of granulocyte colony-stimulating factor of SEQ ID NO:6, wherein said variant contains T133 and wherein a cysteine residue is substituted for at least one amino acid located in at least one region of granulocyte colony-stimulating factor selected from the group consisting of: the A-B loop, the B-C loop, the C-D loop, the first three or last three amino acids in helix B, the first three or last three amino acids in helix C, the first three or last three amino acids in helix D, and the amino acids following helix D; wherein said variant has biological activity in vitro as measured by proliferation of a cell line that proliferates in response to granulocyte colony-stimulating factor.
4. The cysteine variant according to claim 3, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of: K40, S53, G55, W58, A59, P60, S62, S63, P65, S66, Q67, A68, Q70, A72, Q90, A91, E93, G94, S96, E98, G100, G125, M126, A127, A129, Q131, Q134, G135, A136, A139, A141, S142, A143, Q145, Q173 and P174.
5. A cysteine variant of granulocyte colony-stimulating factor of SEQ ID NO:6, wherein a cysteine residue is substituted for at least one amino acid located in the region of granulocyte colony-stimulating factor following helix D; wherein said variant has biological activity in vitro as measured by proliferation of a cell line that proliferates in response to granulocyte colony-stimulating factor.
6. The cysteine variant according to claim 5, wherein a cysteine residue is substituted for Q173.
7. The cysteine variant according to claim 5, wherein a cysteine residue is substituted for P174.
8. A cysteine variant of granulocyte colony-stimulating factor of SEQ ID NO:6, wherein a cysteine residue is substituted for at least one amino acid located in the A-B loop of granulocyte colony-stimulating factor; wherein said variant has biological activity in vitro as measured by proliferation of a cell line that proliferates in response to granulocyte colony-stimulating factor.
9. The cysteine variant according to claim 8, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of K40, S53, G55, W58, A59, P60, S62, S63, P65, S66, Q67, A68, and Q70.
10. The cysteine variant according to claim 8, wherein a cysteine residue is substituted for W58.
11. The cysteine variant according to claim 8, wherein a cysteine residue is substituted for A68.
12. A cysteine variant of granulocyte colony-stimulating factor of SEQ ID NO:6, wherein a cysteine residue is substituted for at least one amino acid located in the B-C loop of granulocyte colony-stimulating factor; wherein said variant has biological activity in vitro as measured by proliferation of a cell line that proliferates in response to granulocyte colony-stimulating factor.
13. The cysteine variant according to claim 12, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of E93, G94, S96, and E98.
14. The cysteine variant according to claim 12, wherein a cysteine residue is substituted for E93.
15. The cysteine variant according to claim 12, wherein a cysteine residue is substituted for E98.
16. A cysteine variant of granulocyte colony-stimulating factor of SEQ ID NO:6, wherein a cysteine residue is substituted for at least one amino acid located in the C-D loop of granulocyte colony-stimulating factor; wherein said variant has biological activity in vitro as measured by proliferation of a cell line that proliferates in response to granulocyte colony-stimulating factor.
17. The cysteine variant according to claim 16, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of G125, M126, A127, A129, Q131, Q134, G135, A136, A139, A141, and S142.
18. The cysteine variant according to claim 16, wherein a cysteine residue is substituted for A129.
19. The cysteine variant according to claim 16, wherein a cysteine residue is substituted for Q131.
20. The cysteine variant according to claim 16, wherein a cysteine residue is substituted for T133.
21. The cysteine variant according to claim 16, wherein a cysteine residue is substituted for Q134.
22. The cysteine variant according to claim 16, wherein a cysteine residue is substituted for A136.
23. The cysteine variant according to claim 16, wherein a cysteine residue is substituted for A139.
24. The cysteine variant according to claim 16, wherein a cysteine residue is substituted for A141.
25. A cysteine variant of granulocyte colony-stimulating factor of SEQ ID NO:6, wherein said variant contains T133 and wherein a cysteine residue is substituted for at least one amino acid located in the C-D loop of granulocyte colony-stimulating factor; wherein said variant has biological activity in vitro as measured by proliferation of a cell line that proliferates in response to granulocyte colony-stimulating factor.
26. The cysteine variant according to claim 25, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of G125, M126, A127, A129, Q131, Q134, G135, A136, A139, A141, and S142.
27. A cysteine variant of granulocyte colony-stimulating factor of SEQ ID NO:6, wherein a cysteine residue is substituted for at least one of the first three amino acids in helix A of granulocyte colony-stimulating factor; wherein said variant has biological activity in vitro as measured by proliferation of a cell line that proliferates in response to granulocyte colony-stimulating factor.
28. The cysteine variant according to claim 27, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of Q11 and S12.
29. A cysteine variant of granulocyte colony-stimulating factor of SEQ ID NO:6, wherein a cysteine residue is substituted for T38; wherein said variant has biological activity in vitro as measured by proliferation of a cell line that proliferates in response to granulocyte colony-stimulating factor.
30. A cysteine variant of granulocyte colony-stimulating factor of SEQ ID NO:6, wherein a cysteine residue is substituted for at least one amino acid located in at least one region of granulocyte colony-stimulating factor selected from the group consisting of the first three amino acids in helix B and the last three amino acids in helix B; wherein said variant has biological activity in vitro as measured by proliferation of a cell line that proliferates in response to granulocyte colony-stimulating factor.
31. The cysteine variant according to claim 30, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of A72, Q90 and A91.
32. A cysteine variant of granulocyte colony-stimulating factor of SEQ ID NO:6, wherein a cysteine residue is substituted for at least one amino acid located in at least one region of granulocyte colony-stimulating factor selected from the group consisting of the first three amino acids in helix C and the last three amino acids in helix C; wherein said variant has biological activity in vitro as measured by proliferation of a cell line that proliferates in response to granulocyte colony-stimulating factor.
33. The cysteine variant according to claim 33, wherein a cysteine residue is substituted for G100.
34. A cysteine variant of granulocyte colony-stimulating factor of SEQ ID NO:6, wherein a cysteine residue is substituted for at least one amino acid located in at least one region of granulocyte colony-stimulating factor selected from the group consisting of the first three amino acids in helix D and the last three amino acids in helix D; wherein said variant has biological activity in vitro as measured by proliferation of a cell line that proliferates in response to granulocyte colony-stimulating factor.
35. The cysteine variant according to claim 34, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of A143 and Q145.
36. A cysteine variant of granulocyte colony-stimulating factor of SEQ ID NO:6, wherein a cysteine residue is substituted for at least one amino acid of granulocyte colony-stimulating factor selected from the group consisting of P2, S7, S8, L9 and P10; wherein said variant has biological activity in vitro as measured by proliferation of a cell line that proliferates in response to granulocyte colony-stimulating factor.
37. The cysteine variant according to claim 36, wherein a cysteine residue is substituted for P2.
38. The cysteine variant according to claim 36, wherein a cysteine residue is substituted for S7.
39. The cysteine variant according to any one of claims 1, 3, 5, 8, 12, 16, 25, 27, 29, 30, 32, 34 or 36, wherein a non-cysteine amino acid is substituted for C17.
40. The cysteine variant according to claim 39, wherein the non-cysteine amino acid substituted for C17 is selected from the group consisting of serine and alanine.
41. The cysteine variant according to any one of claims 1, 3, 5, 8, 12, 16, 25, 27, 29, 30, 32, 34 or 36, wherein the substituted cysteine residue is modified with a cysteine-reactive moiety.
42. The cysteine variant according to any one of claims 1, 3, 5, 8, 12, 16, 25, 27, 29, 30, 32, 34 or 36, wherein the substituted cysteine residue is modified with a cysteine-reactive moiety, and wherein a non-cysteine amino acid is substituted for C17.
43. The cysteine variant according to claim 42, wherein the non-cysteine amino acid substituted for C17 is selected from the group consisting of serine and alanine.
44. The cysteine variant according to any one of claims 1, 3, 5, 8, 12, 16, 25, 27, 29, 30, 32, 34 or 36, wherein the substituted cysteine residue is modified with polyethylene glycol.
45. The cysteine variant according to any one of claims 1, 3, 5, 8, 12, 16, 25, 27, 29, 30, 32, 34 or 36, wherein the substituted cysteine residue is modified with polyethylene glycol, and wherein a non-cysteine amino acid is substituted for C17.
46. The cysteine variant according to claim 45, wherein the non-cysteine amino acid substituted for C17 is selected from the group consisting of serine and alanine.
47. The cysteine variant according to any one of claims 1, 3, 5, 8, 12, 16, 25, 27, 29, 30, 32, 34 or 36, wherein said cysteine variant is modified with at least one polyethylene glycol.
48. The cysteine variant according to any one of claims 1, 3, 5, 8, 12, 16, 25, 27, 29, 30, 32, 34 or 36, wherein said cysteine variant is modified with at least one polyethylene glycol, and wherein a non-cysteine amino acid is substituted for C17.
49. The cysteine variant according to claim 48, wherein the non-cysteine amino acid substituted for C17 is selected from the group consisting of serine and alanine.
50. A cysteine variant of granulocyte colony-stimulating factor of SEQ ID NO:6, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of T1, L3, G4, P5, and A6, and wherein the substituted cysteine residue is modified with a cysteine-reactive moiety; wherein said variant has biological activity in vitro as measured by proliferation of a cell line that proliferates in response to granulocyte colony-stimulating factor.
51. The cysteine variant according to claim 50, wherein a cysteine residue is substituted for T1.
52. The cysteine variant according to claim 50, wherein a cysteine residue is substituted for L3.
53. The cysteine variant according to claim 50, wherein a cysteine residue is substituted for A6.
54. A cysteine variant of granulocyte colony-stimulating factor of SEQ ID NO:6, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of T1, L3, G4, P5, and A6, and wherein the substituted cysteine residue is modified with a polyethylene glycol; wherein said variant has biological activity in vitro as measured by proliferation of a cell line that proliferates in response to granulocyte colony-stimulating factor.
55. The cysteine variant according to claim 54, wherein a cysteine residue is substituted for T1.
56. The cysteine variant according to claim 54, wherein a cysteine residue is substituted for L3.
57. The cysteine variant according to claim 54, wherein a cysteine residue is substituted for A6.
58. The cysteine variant according to any one of claims 50 or 53, wherein a non-cysteine amino acid is substituted for C17.
59. The cysteine variant according to claim 58, wherein the non-cysteine amino acid substituted for C17 is selected from the group consisting of serine and alanine.
US10/856,219 1997-07-14 2004-05-27 Cysteine variants of granulocyte colony-stimulating factor Expired - Fee Related US7309781B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/856,219 US7309781B2 (en) 1997-07-14 2004-05-27 Cysteine variants of granulocyte colony-stimulating factor

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US5251697P 1997-07-14 1997-07-14
US09/462,941 US6608183B1 (en) 1997-07-14 1998-07-13 Derivatives of growth hormone and related proteins
PCT/US1998/014497 WO1999003887A1 (en) 1997-07-14 1998-07-13 Derivatives of growth hormone and related proteins
US10/400,377 US7148333B2 (en) 1997-07-14 2003-03-26 Cysteine variants of granulocyte-macrophage colony-stimulating factor
US10/856,219 US7309781B2 (en) 1997-07-14 2004-05-27 Cysteine variants of granulocyte colony-stimulating factor

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/400,377 Continuation US7148333B2 (en) 1997-07-14 2003-03-26 Cysteine variants of granulocyte-macrophage colony-stimulating factor

Publications (2)

Publication Number Publication Date
US20040265269A1 true US20040265269A1 (en) 2004-12-30
US7309781B2 US7309781B2 (en) 2007-12-18

Family

ID=21978119

Family Applications (18)

Application Number Title Priority Date Filing Date
US09/462,941 Expired - Lifetime US6608183B1 (en) 1997-07-14 1998-07-13 Derivatives of growth hormone and related proteins
US10/400,708 Abandoned US20030166865A1 (en) 1997-07-14 2003-03-26 Cysteine variants of interleukin-11
US10/400,377 Expired - Lifetime US7148333B2 (en) 1997-07-14 2003-03-26 Cysteine variants of granulocyte-macrophage colony-stimulating factor
US10/774,149 Expired - Fee Related US7232885B2 (en) 1997-07-14 2004-02-05 Termini cysteine-added variants of granulocyte colony stimulating factor
US10/773,939 Expired - Lifetime US7253267B2 (en) 1997-07-14 2004-02-05 Cysteine variants of interleukin-11
US10/773,530 Expired - Fee Related US7345154B2 (en) 1997-07-14 2004-02-05 Cysteine variants of erythropoietin
US10/773,654 Expired - Fee Related US7214779B2 (en) 1997-07-14 2004-02-05 Termini cysteine-added variants of granulocyte-macrophage colony stimulating factor
US10/856,219 Expired - Fee Related US7309781B2 (en) 1997-07-14 2004-05-27 Cysteine variants of granulocyte colony-stimulating factor
US10/866,540 Expired - Fee Related US7732572B2 (en) 1997-07-14 2004-06-10 Cysteine variants of alpha interferon-2
US10/866,580 Expired - Fee Related US7314921B2 (en) 1997-07-14 2004-06-10 Cysteine variants of erythropoietin
US11/071,098 Expired - Fee Related US7795396B2 (en) 1997-07-14 2005-03-02 COOH-terminally added cysteine variant of the beta interferon
US11/070,993 Expired - Fee Related US7345149B2 (en) 1997-07-14 2005-03-02 Cysteine substitution variants of beta interferon
US11/929,521 Expired - Fee Related US7959909B2 (en) 1997-07-14 2007-10-30 Cysteine variants of interferon gamma
US11/929,504 Abandoned US20090060863A1 (en) 1997-07-14 2007-10-30 Derivatives of growth hormone and related proteins
US11/929,478 Expired - Fee Related US7964184B2 (en) 1997-07-14 2007-10-30 Cysteine variants of interferon-gamma
US13/107,021 Expired - Fee Related US8618256B2 (en) 1997-07-14 2011-05-13 Cysteine variants of interferon gamma
US14/089,498 Abandoned US20140163204A1 (en) 1997-07-14 2013-11-25 Derivatives of growth hormone and related proteins
US15/470,041 Active US10329337B2 (en) 1997-07-14 2017-03-27 Method to increase the number of circulating platelets by administering PEGylated cysteine variants of IL-11

Family Applications Before (7)

Application Number Title Priority Date Filing Date
US09/462,941 Expired - Lifetime US6608183B1 (en) 1997-07-14 1998-07-13 Derivatives of growth hormone and related proteins
US10/400,708 Abandoned US20030166865A1 (en) 1997-07-14 2003-03-26 Cysteine variants of interleukin-11
US10/400,377 Expired - Lifetime US7148333B2 (en) 1997-07-14 2003-03-26 Cysteine variants of granulocyte-macrophage colony-stimulating factor
US10/774,149 Expired - Fee Related US7232885B2 (en) 1997-07-14 2004-02-05 Termini cysteine-added variants of granulocyte colony stimulating factor
US10/773,939 Expired - Lifetime US7253267B2 (en) 1997-07-14 2004-02-05 Cysteine variants of interleukin-11
US10/773,530 Expired - Fee Related US7345154B2 (en) 1997-07-14 2004-02-05 Cysteine variants of erythropoietin
US10/773,654 Expired - Fee Related US7214779B2 (en) 1997-07-14 2004-02-05 Termini cysteine-added variants of granulocyte-macrophage colony stimulating factor

Family Applications After (10)

Application Number Title Priority Date Filing Date
US10/866,540 Expired - Fee Related US7732572B2 (en) 1997-07-14 2004-06-10 Cysteine variants of alpha interferon-2
US10/866,580 Expired - Fee Related US7314921B2 (en) 1997-07-14 2004-06-10 Cysteine variants of erythropoietin
US11/071,098 Expired - Fee Related US7795396B2 (en) 1997-07-14 2005-03-02 COOH-terminally added cysteine variant of the beta interferon
US11/070,993 Expired - Fee Related US7345149B2 (en) 1997-07-14 2005-03-02 Cysteine substitution variants of beta interferon
US11/929,521 Expired - Fee Related US7959909B2 (en) 1997-07-14 2007-10-30 Cysteine variants of interferon gamma
US11/929,504 Abandoned US20090060863A1 (en) 1997-07-14 2007-10-30 Derivatives of growth hormone and related proteins
US11/929,478 Expired - Fee Related US7964184B2 (en) 1997-07-14 2007-10-30 Cysteine variants of interferon-gamma
US13/107,021 Expired - Fee Related US8618256B2 (en) 1997-07-14 2011-05-13 Cysteine variants of interferon gamma
US14/089,498 Abandoned US20140163204A1 (en) 1997-07-14 2013-11-25 Derivatives of growth hormone and related proteins
US15/470,041 Active US10329337B2 (en) 1997-07-14 2017-03-27 Method to increase the number of circulating platelets by administering PEGylated cysteine variants of IL-11

Country Status (14)

Country Link
US (18) US6608183B1 (en)
EP (2) EP1881005B1 (en)
JP (2) JP2001510033A (en)
CN (1) CN1269805A (en)
AT (1) ATE375363T1 (en)
AU (1) AU751898B2 (en)
BR (1) BR9812267B1 (en)
CA (1) CA2296770A1 (en)
DE (1) DE69838552T2 (en)
ES (1) ES2297889T3 (en)
HK (2) HK1028617A1 (en)
IL (3) IL133974A0 (en)
NZ (1) NZ502375A (en)
WO (1) WO1999003887A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080075672A1 (en) * 2002-09-09 2008-03-27 Rene Gantier Rational evolution of cytokines for higher stability, the cytokines and encoding nucleic acid molecules
US20080076706A1 (en) * 1997-07-14 2008-03-27 Bolder Biotechnology, Inc. Derivatives of Growth Hormone and Related Proteins, and Methods of Use Thereof
WO2009139784A1 (en) * 2008-05-13 2009-11-19 Nora Therapeutics, Inc. Human g-csf analogs and methods of making and using thereof
US20110171168A1 (en) * 2008-05-13 2011-07-14 Nora Therapeutics, Inc. Human g-csf analogs and methods of making and using thereof

Families Citing this family (312)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9526733D0 (en) * 1995-12-30 1996-02-28 Delta Biotechnology Ltd Fusion proteins
US6753165B1 (en) * 1999-01-14 2004-06-22 Bolder Biotechnology, Inc. Methods for making proteins containing free cysteine residues
US7495087B2 (en) 1997-07-14 2009-02-24 Bolder Biotechnology, Inc. Cysteine muteins in the C-D loop of human interleukin-11
US7153943B2 (en) 1997-07-14 2006-12-26 Bolder Biotechnology, Inc. Derivatives of growth hormone and related proteins, and methods of use thereof
US7306931B2 (en) 2000-05-16 2007-12-11 Bolder Biotechnology, Inc. Method for refolding proteins containing free cysteine residues
EP1881005B1 (en) * 1997-07-14 2013-04-03 Bolder Biotechnology, Inc. Derivatives of G-CSF and related proteins
US7270809B2 (en) * 1997-07-14 2007-09-18 Bolder Biotechnology, Inc. Cysteine variants of alpha interferon-2
US6747003B1 (en) 1997-10-23 2004-06-08 Regents Of The University Of Minnesota Modified vitamin K-dependent polypeptides
PL193352B1 (en) * 1998-04-28 2007-02-28 Applied Research Systems Polyol-ifn-beta conjugates
IL142350A0 (en) 1998-10-16 2002-03-10 Biogen Inc Interferon-beta fusion proteins and pharmaceutical compositions containing the same
SG2008070138A (en) 1998-10-16 2017-08-30 Biogen Ma Inc Polymer conjugates of interferon beta- 1a and their uses
US8288126B2 (en) 1999-01-14 2012-10-16 Bolder Biotechnology, Inc. Methods for making proteins containing free cysteine residues
BR0008759B1 (en) * 1999-01-14 2014-03-11 Bolder Biotechnology Inc Methods for the production of protein containing free cysteine residues
US7431921B2 (en) 2000-04-14 2008-10-07 Maxygen Aps Interferon beta-like molecules
US6531122B1 (en) 1999-08-27 2003-03-11 Maxygen Aps Interferon-β variants and conjugates
US7144574B2 (en) 1999-08-27 2006-12-05 Maxygen Aps Interferon β variants and conjugates
IL147581A0 (en) * 1999-08-27 2002-08-14 Maxygen Aps New interferon beta-like molecules
AU2154401A (en) * 1999-11-12 2001-05-30 Merck Patent Gmbh Erythropoietin forms with improved properties
KR20020065517A (en) 1999-11-12 2002-08-13 맥시겐 홀딩스 리미티드 Interferon gamma conjugates
AU782635B2 (en) * 1999-11-12 2005-08-18 Maxygen Holdings Ltd. Interferon gamma conjugates
US7230081B1 (en) 1999-11-12 2007-06-12 Maxygen Holdings, Ltd. Interferon gamma conjugates
US6831158B2 (en) 2000-01-10 2004-12-14 Maxygen Holdings Ltd. G-CSF conjugates
US6555660B2 (en) 2000-01-10 2003-04-29 Maxygen Holdings Ltd. G-CSF conjugates
WO2001058950A1 (en) * 2000-02-11 2001-08-16 Maxygen Aps Improved interleukin 10
IL150692A0 (en) * 2000-02-11 2003-02-12 Maxygen Aps FACTOR VII OR VIIa-LIKE MOLECULES
AU2001238595A1 (en) 2000-02-22 2001-09-03 Shearwater Corporation N-maleimidyl polymer derivatives
US7812132B2 (en) 2000-04-28 2010-10-12 Regents Of The University Of Minnesota Modified vitamin K-dependent polypeptides
US7220837B1 (en) 2000-04-28 2007-05-22 Regents Of The University Of Minnesota Modified vitamin K-dependent polypeptides
US20030211094A1 (en) 2001-06-26 2003-11-13 Nelsestuen Gary L. High molecular weight derivatives of vitamin k-dependent polypeptides
US7118737B2 (en) * 2000-09-08 2006-10-10 Amylin Pharmaceuticals, Inc. Polymer-modified synthetic proteins
AU7338801A (en) * 2000-09-08 2002-03-22 Gryphon Sciences "pseudo"-native chemical ligation
ES2320101T3 (en) * 2000-10-16 2009-05-19 Chugai Seiyaku Kabushiki Kaisha CONJUGATED ERYTHROPOYETINE WITH MONO-PEG.
US6673580B2 (en) 2000-10-27 2004-01-06 Genentech, Inc. Identification and modification of immunodominant epitopes in polypeptides
EP1334128A2 (en) * 2000-11-02 2003-08-13 Maxygen Aps New multimeric interferon beta polypeptides
EP2062593A3 (en) 2000-12-01 2011-08-17 Takeda Pharmaceutical Company Limited Method for producing preparation containing bioactive peptide
EP1352062A2 (en) * 2001-01-11 2003-10-15 Maxygen Aps Improved growth hormone molecules
US7442370B2 (en) 2001-02-01 2008-10-28 Biogen Idec Ma Inc. Polymer conjugates of mutated neublastin
RU2003125654A (en) * 2001-02-06 2005-03-10 Мерк Патент ГмбХ (DE) MODIFIED ERYTHROPOETIN (EPO) WITH REDUCED IMMUNOGENICITY
MXPA03007619A (en) 2001-02-27 2003-12-04 Maxygen Aps New interferon beta-like molecules.
GB0105360D0 (en) * 2001-03-03 2001-04-18 Glaxo Group Ltd Chimaeric immunogens
US6992174B2 (en) 2001-03-30 2006-01-31 Emd Lexigen Research Center Corp. Reducing the immunogenicity of fusion proteins
US7038015B2 (en) 2001-04-06 2006-05-02 Maxygen Holdings, Ltd. Interferon gamma polypeptide variants
US6958388B2 (en) 2001-04-06 2005-10-25 Maxygen, Aps Interferon gamma polypeptide variants
JP4123856B2 (en) 2001-07-31 2008-07-23 日油株式会社 Bio-related substance modifier and method for producing polyoxyalkylene derivative
JP2005508162A (en) 2001-10-02 2005-03-31 ジェネンテック・インコーポレーテッド Apo-2 ligand variants and methods of use
US6908963B2 (en) 2001-10-09 2005-06-21 Nektar Therapeutics Al, Corporation Thioester polymer derivatives and method of modifying the N-terminus of a polypeptide therewith
US20030171285A1 (en) * 2001-11-20 2003-09-11 Finn Rory F. Chemically-modified human growth hormone conjugates
EA008505B1 (en) * 2001-11-20 2007-06-29 Фармация Корпорейшн Chemically modified human growth hormone conjugates
KR100467750B1 (en) * 2001-11-29 2005-01-24 씨제이 주식회사 Fusion protein having the enhanced in vivo activity of erythropoietin
US7247609B2 (en) 2001-12-18 2007-07-24 Universitat Zurich Growth factor modified protein matrices for tissue engineering
US20080167238A1 (en) * 2001-12-21 2008-07-10 Human Genome Sciences, Inc. Albumin Fusion Proteins
WO2003055526A2 (en) * 2001-12-21 2003-07-10 Maxygen Aps Erythropoietin conjugates
WO2003064462A1 (en) * 2002-02-01 2003-08-07 Chugai Seiyaku Kabushiki Kaisha Peg-binding pth or peg-binding pth derivative
EP1499719B1 (en) 2002-04-30 2010-11-17 Bayer HealthCare LLC FACTOR VII OR VIIa POLYPEPTIDE VARIANTS
CA2489348A1 (en) 2002-06-24 2003-12-31 Genentech, Inc. Apo-2 ligand/trail variants and uses thereof
WO2004005341A2 (en) * 2002-07-03 2004-01-15 Maxygen Holdings Ltd. Full-length interferon gamma polypeptide variants
EP1391513A1 (en) 2002-08-08 2004-02-25 Cytheris IL-7 drug substance, IL-7 comprising composition, preparation and uses thereof
US20040121953A1 (en) * 2002-08-09 2004-06-24 Chirino Arthur J. Thrombopoiesis-stimulating proteins having reduced immunogenicity
WO2004020468A2 (en) * 2002-08-28 2004-03-11 Maxygen Aps Interferon beta-like molecules for treatment of cancer
US8129330B2 (en) 2002-09-30 2012-03-06 Mountain View Pharmaceuticals, Inc. Polymer conjugates with decreased antigenicity, methods of preparation and uses thereof
WO2004044006A1 (en) * 2002-11-14 2004-05-27 Maxygen, Inc. Conjugates of interleukin-10 and polymers
US7314613B2 (en) 2002-11-18 2008-01-01 Maxygen, Inc. Interferon-alpha polypeptides and conjugates
MXPA05006944A (en) 2002-12-26 2005-12-14 Mountain View Pharmaceuticals Polymer conjugates of interferon-beta.
RS20050501A (en) * 2002-12-26 2007-08-03 Mountain View Pharmaceuticals Inc., Polymer conjugates of cytokines,chemokines,growth factors, polypeptide hormones and antagonists thereof with preserved receptor-binding activity
US7432331B2 (en) 2002-12-31 2008-10-07 Nektar Therapeutics Al, Corporation Hydrolytically stable maleimide-terminated polymers
AU2003300133B2 (en) 2002-12-31 2008-11-13 Nektar Therapeutics Hydrolytically stable maleimide-terminated polymers
KR101031206B1 (en) 2002-12-31 2011-04-27 넥타르 테라퓨틱스 Maleamic acid polymer derivatives and their bioconjugates
CN102367277B (en) * 2003-01-31 2014-04-09 比奥根艾迪克Ma公司 Polymer conjugates of mutated neublastin
DE602004021099D1 (en) 2003-03-20 2009-06-25 Bayer Healthcare Llc FVII OR FVIIA VARIANTS
US20070184519A1 (en) * 2003-04-02 2007-08-09 Shabnam Tangri Peptides, polypeptides, and proteins of reduced immunogenicity and methods for their production
WO2004091517A2 (en) 2003-04-15 2004-10-28 Smithkline Beecham Corporation Conjugates comprising human il-18 and substitution mutants thereof
WO2004099245A1 (en) * 2003-05-05 2004-11-18 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Stabilised interferon-ϝ
BRPI0411650A (en) 2003-06-19 2006-08-08 Maxygen Holdings Ltd polypeptide variant of factor vii (fvii) or factor viia (fviia), nucleotide sequence, expression vector, host cell, composition, use of a variant, and method for treating a mammal having a disease or disorder in which clot formation is desirable
US7785580B2 (en) 2003-08-29 2010-08-31 Aerovance, Inc. Modified IL-4 mutein receptor antagonists
US7404957B2 (en) * 2003-08-29 2008-07-29 Aerovance, Inc. Modified IL-4 mutein receptor antagonists
EP2263684A1 (en) * 2003-10-10 2010-12-22 Novo Nordisk A/S IL-21 derivatives
US20060228331A1 (en) 2003-10-10 2006-10-12 Novo Nordisk A/S IL-21 Derivatives and variants
US7790835B2 (en) 2003-12-03 2010-09-07 Nektar Therapeutics Method of preparing maleimide functionalized polymers
AU2004311796A1 (en) * 2003-12-31 2005-07-21 Centocor, Inc. Novel recombinant proteins with N-terminal free thiol
WO2005070967A2 (en) 2004-01-22 2005-08-04 Merck Patent Gmbh Anti-cancer antibodies with reduced complement fixation
CN103755800B (en) * 2004-02-02 2016-02-24 Ambrx公司 Modified human interferon polypeptides and its purposes
US7588745B2 (en) * 2004-04-13 2009-09-15 Si Options, Llc Silicon-containing products
EP1586334A1 (en) * 2004-04-15 2005-10-19 TRASTEC scpa G-CSF conjugates with peg
JP2008507298A (en) 2004-05-19 2008-03-13 マキシジェン, インコーポレイテッド Interferon alpha polypeptides and conjugates
CN102603895B (en) 2004-06-18 2016-09-28 Ambrx公司 Novel antigen-binding polypeptides and its purposes
CA2573262C (en) * 2004-07-16 2015-02-10 Nektar Therapeutics Al, Corporation Conjugates of a gm-csf moiety and a polymer
ES2349043T3 (en) 2004-08-19 2010-12-22 Biogen Idec Ma Inc. NEUBLASTIN VARIANTS.
MX2007002441A (en) * 2004-08-31 2007-05-04 Pharmacia & Upjohn Co Llc Glycerol branched polyethylene glycol human growth hormone conjugates, process for their preparation, and methods of use thereof.
US7998930B2 (en) 2004-11-04 2011-08-16 Hanall Biopharma Co., Ltd. Modified growth hormones
EP2363414B1 (en) 2004-11-12 2022-05-18 Bayer HealthCare LLC Site-directed modification of FVIII
GB2438982A (en) * 2004-12-22 2007-12-12 Ambrx Inc Compostions containing, methods involving, and uses of non-natural amino acids and polypeptides
AU2005319518B2 (en) * 2004-12-22 2010-09-09 Ambrx, Inc. Compositions of aminoacyl-tRNA synthetase and uses thereof
US7816320B2 (en) 2004-12-22 2010-10-19 Ambrx, Inc. Formulations of human growth hormone comprising a non-naturally encoded amino acid at position 35
SG160437A1 (en) 2004-12-22 2010-04-29 Ambrx Inc Modified human growth hormone
JP2008525032A (en) 2004-12-22 2008-07-17 アンブレツクス・インコーポレイテツド Methods for expressing and purifying recombinant human growth hormone
US7714114B2 (en) 2005-02-16 2010-05-11 Nektar Therapeutics Conjugates of an EPO moiety and a polymer
WO2007044083A2 (en) 2005-05-18 2007-04-19 Maxygen, Inc. Evolved interferon-alpha polypeptides
CA2607844C (en) 2005-06-01 2012-07-10 Maxygen Holdings Ltd. Pegylated g-csf polypeptides and methods of producing same
CA2609205A1 (en) * 2005-06-03 2006-12-14 Ambrx, Inc. Improved human interferon molecules and their uses
KR100694994B1 (en) * 2005-06-13 2007-03-14 씨제이 주식회사 Human Granulocyte-Colony Stimulating Factor Isoforms
CA2613737C (en) 2005-06-29 2017-05-23 Yeda Research And Development Co. Ltd. Recombinant interferon .alpha.2 (ifn.alpha.2) mutants
KR100735784B1 (en) * 2005-07-20 2007-07-06 재단법인 목암생명공학연구소 Mutant of granulocyte-colony stimulating factorG-CSF and chemically conjugated polypeptide thereof
EP1951890A4 (en) 2005-11-16 2009-06-24 Ambrx Inc Methods and compositions comprising non-natural amino acids
NZ568578A (en) 2005-12-14 2011-10-28 Ambrx Inc Compositions containing, methods involving, and uses of non-natural amino acids and polypeptides
US7659250B2 (en) 2005-12-22 2010-02-09 Centocor, Inc. BMP-7 variant compositions, methods and uses
EP1978994B1 (en) * 2005-12-22 2012-02-01 Janssen Biotech, Inc. Bmp-7 variant compositions, methods and uses
SI1966238T1 (en) 2005-12-30 2012-08-31 Merck Patent Gmbh Interleukin-12p40 variants with improved stability
PT2270050E (en) 2005-12-30 2013-09-12 Merck Patent Gmbh Anti-cd19 antibodies with reduced immunogenicity
ES2546282T3 (en) * 2006-02-14 2015-09-22 Novo Nordisk Health Care Ag Polypeptide coupling at the C-terminal end
TWI501774B (en) 2006-02-27 2015-10-01 Biogen Idec Inc Treatments for neurological disorders
US20080038224A1 (en) * 2006-03-28 2008-02-14 Thierry Guyon Modified interferon-beta (IFN-beta) polypeptides
CN101495536B (en) 2006-05-26 2012-03-07 益普生制药股份有限公司 Methods for site-specific pegylation
JP2009544325A (en) * 2006-07-27 2009-12-17 セントカー・インコーポレーテツド BMP-7 variant compositions, methods and uses
PT2061878E (en) 2006-09-08 2014-04-22 Ambrx Inc Hybrid suppressor trna for vertebrate cells
WO2008030558A2 (en) 2006-09-08 2008-03-13 Ambrx, Inc. Modified human plasma polypeptide or fc scaffolds and their uses
NZ574960A (en) 2006-09-08 2012-02-24 Ambrx Inc Suppressor trna transcription in vertebrate cells
WO2008049920A2 (en) 2006-10-26 2008-05-02 Novo Nordisk A/S Il-21 variants
JP2010510794A (en) * 2006-11-28 2010-04-08 ハナル ファーマシューティカル カンパニー リミテッド Modified erythropoietin polypeptide and therapeutic use thereof
EP2102355B1 (en) 2006-12-14 2016-03-02 Bolder Biotechnology, Inc. Long acting proteins and peptides and methods of making and using the same
PE20090722A1 (en) * 2007-02-02 2009-07-13 Amgen Inc HEPCIDIN, HEPCIDIN ANTAGONISTS AND METHODS OF USE
CA2682147C (en) 2007-03-30 2017-08-08 Ambrx, Inc. Modified fgf-21 polypeptides and their uses
ES2476253T3 (en) 2007-05-01 2014-07-14 Biogen Idec Ma Inc. Neublastin peptides for use in increasing vascularization in tissue with impaired blood flow
BRPI0810622A2 (en) 2007-05-02 2020-10-13 Ambrx, Inc. modified interferon beta polypeptides and their uses
MX2009013259A (en) 2007-06-12 2010-01-25 Novo Nordisk As Improved process for the production of nucleotide sugars.
AU2008304111B2 (en) * 2007-09-27 2014-04-24 Amgen Inc. Pharmaceutical formulations
US8796206B2 (en) 2007-11-15 2014-08-05 Amgen Inc. Aqueous formulation of erythropoiesis stimulating protein stabilised by antioxidants for parenteral administration
JP5547083B2 (en) 2007-11-20 2014-07-09 アンブルックス,インコーポレイテッド Modified insulin polypeptides and their use
US20110124841A1 (en) * 2007-12-13 2011-05-26 Monthony James F Polypeptides Modified by Protein Trans-Splicing Technology
CA2710798A1 (en) 2007-12-28 2009-07-09 Kuros Biosurgery Ag Pdgf fusion proteins incorporated into fibrin foams
US8101706B2 (en) * 2008-01-11 2012-01-24 Serina Therapeutics, Inc. Multifunctional forms of polyoxazoline copolymers and drug compositions comprising the same
EP3042922B1 (en) 2008-01-11 2017-07-19 Serina Therapeutics, Inc. Multifunctional forms of polyoxazoline copolymers and drug compositions comprising the same
WO2009094551A1 (en) 2008-01-25 2009-07-30 Amgen Inc. Ferroportin antibodies and methods of use
MX2010008632A (en) 2008-02-08 2010-08-30 Ambrx Inc Modified leptin polypeptides and their uses.
CN101525381B (en) * 2008-03-04 2012-04-18 北京百川飞虹生物科技有限公司 Novel recombinant consensus interferon and construction of a high-efficiency expression vector thereof
JP2011519279A (en) 2008-05-01 2011-07-07 アムジエン・インコーポレーテツド Anti-hepcidin antibodies and methods of use
NZ591235A (en) 2008-07-23 2012-07-27 Ambrx Inc Modified bovine g-csf polypeptides comprising non natural amino acid and their uses treating infections such as mastitis
MX2011000847A (en) * 2008-08-06 2011-02-25 Novo Nordisk Healthcare Ag Conjugated proteins with prolonged in vivo efficacy.
NO2337846T3 (en) 2008-09-26 2018-06-16
NZ592250A (en) 2008-09-26 2012-11-30 Lilly Co Eli Modified animal erythropoietin polypeptides and their uses
CN104922669A (en) 2008-11-13 2015-09-23 通用医疗公司 Methods and compositions for regulating iron homeostasis by modulation bmp-6
WO2010064132A1 (en) * 2008-12-05 2010-06-10 Council Of Scientific & Industrial Research Mutants of staphylokinase carrying amino and carboxy-terminal extensions for polyethylene glycol conjugation
EP2389389B1 (en) * 2009-01-22 2015-04-15 Novo Nordisk Health Care AG Stable growth hormone compounds
US8940501B2 (en) 2009-01-30 2015-01-27 Whitehead Institute For Biomedical Research Methods for ligation and uses thereof
US9238878B2 (en) 2009-02-17 2016-01-19 Redwood Bioscience, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
EP2421896A1 (en) 2009-04-22 2012-02-29 Merck Patent GmbH Antibody fusion proteins with modified fcrn binding sites
JP5751641B2 (en) 2009-05-06 2015-07-22 ヤンセン バイオテツク,インコーポレーテツド Melanocortin receptor binding conjugate
CN105963710A (en) * 2009-08-06 2016-09-28 诺沃—诺迪斯克保健股份有限公司 Growth hormones with prolonged in-vivo efficacy
US20120178914A1 (en) * 2009-09-25 2012-07-12 Vybion, Inc. Polypeptide modification
MX344382B (en) 2009-10-23 2016-12-14 Amgen Inc * Vial adapter and system.
KR20120123299A (en) 2009-12-04 2012-11-08 제넨테크, 인크. Multispecific antibodies, antibody analogs, compositions, and methods
CN107674121A (en) 2009-12-21 2018-02-09 Ambrx 公司 Bovine somatotropin polypeptide and its purposes by modification
CN104017063A (en) 2009-12-21 2014-09-03 Ambrx公司 Modified porcine somatotropin polypeptides and their uses
RS59459B1 (en) * 2010-01-22 2019-11-29 Novo Nordisk Healthcare Ag Growth hormones with prolonged in-vivo efficacy
AU2011208620B2 (en) 2010-01-22 2015-04-16 Novo Nordisk Health Care Ag Stable growth hormone compounds
WO2011133948A2 (en) 2010-04-22 2011-10-27 Longevity Biotech, Inc. Highly active polypeptides and methods of making and using the same
HUE026173T2 (en) 2010-06-07 2016-05-30 Amgen Inc Drug delivery device
WO2012010516A1 (en) * 2010-07-22 2012-01-26 Novo Nordisk Health Care Ag Growth hormone conjugates
US9567386B2 (en) 2010-08-17 2017-02-14 Ambrx, Inc. Therapeutic uses of modified relaxin polypeptides
WO2012024452A2 (en) 2010-08-17 2012-02-23 Ambrx, Inc. Modified relaxin polypeptides and their uses
TWI480288B (en) 2010-09-23 2015-04-11 Lilly Co Eli Formulations for bovine granulocyte colony stimulating factor and variants thereof
CA2824143C (en) 2011-01-14 2018-12-18 Redwood Bioscience, Inc. Aldehyde-tagged immunoglobulin polypeptides and method of use thereof
WO2012099886A1 (en) 2011-01-18 2012-07-26 Bioniz, Llc Compositions and mehthods for modulating gamma-c-cytokine activity
CA2826114A1 (en) * 2011-02-01 2012-08-09 Fate Therapeutics, Inc. Cardiotrophin related molecules for enhanced therapeutics
CA2831100C (en) 2011-03-31 2020-02-18 Mark Dominis Holt Vial adapter and system
EP2694095B1 (en) 2011-04-05 2018-03-07 Longevity Biotech, Inc. Compositions comprising glucagon analogs and methods of making and using the same
US10092706B2 (en) 2011-04-20 2018-10-09 Amgen Inc. Autoinjector apparatus
US9320777B2 (en) 2011-05-13 2016-04-26 Bolder Biotechnology, Inc. Methods and use of growth hormone supergene family protein analogs for treatment of radiation exposure
JP2014520094A (en) * 2011-05-27 2014-08-21 バクスター・インターナショナル・インコーポレイテッド Therapeutic proteins with increased half-life and methods for preparing the same
WO2013003555A1 (en) 2011-06-28 2013-01-03 Whitehead Institute For Biomedical Research Using sortases to install click chemistry handles for protein ligation
CA3175560A1 (en) 2011-10-14 2013-04-18 Amgen Inc. Injector and method of assembly
BR112014009925B1 (en) 2011-10-28 2022-09-20 Teva Pharmaceuticals Australia Pty Ltd POLYPEPTIDE BUILDERS AND THEIR USES
EP3505534A1 (en) 2012-06-08 2019-07-03 Sutro Biopharma, Inc. Antibodies comprising sitespecific nonnatural amino acid residues, methods of their preparation and methods of their use
CN103509102B (en) * 2012-06-15 2015-07-22 郭怀祖 Wild type human growth hormone mutant
DK2863955T3 (en) 2012-06-26 2017-01-23 Sutro Biopharma Inc MODIFIED FC PROTEINS, INCLUDING LOCATION-SPECIFIC NON-NATURAL AMINO ACID RESIDUES, CONJUGATES THEREOF, METHODS OF PRODUCING ITS AND PROCEDURES FOR USE THEREOF
BR112015004022B1 (en) 2012-08-31 2023-04-25 Sutro Biopharma, Inc MODIFIED AMINO ACIDS COMPRISING AN AZID GROUP
GB2507341A (en) * 2012-10-29 2014-04-30 Peter Kenny Fat loss composition comprising a prostaglandin F2 alpha analogue and a fragment of amino acids 176-191 of human growth hormone
AU2013348071B2 (en) 2012-11-21 2018-05-24 Amgen Inc. Drug delivery device
EP2958584A1 (en) 2013-02-20 2015-12-30 Yeda Research and Development Co., Ltd. Interferon treatment targeting mutant p53 expressing cells
WO2014139994A1 (en) 2013-03-11 2014-09-18 Novo Nordisk Health Care Ag Growth hormone compounds
WO2014145718A2 (en) 2013-03-15 2014-09-18 Longevity Biotech, Inc. Peptides comprising non-natural amino acids and methods of making and using the same
CA2904725C (en) 2013-03-15 2022-04-12 Amgen Inc. Drug cassette, autoinjector, and autoinjector system
DK2968503T3 (en) 2013-03-15 2018-12-03 Intrinsic Lifesciences Llc ANTI-HEPCIDIN ANTIBODIES AND APPLICATIONS THEREOF
CA2904661C (en) 2013-03-15 2022-03-15 Amgen Inc. Drug cassette, autoinjector, and autoinjector system
MX2015013533A (en) 2013-03-22 2016-02-05 Amgen Inc Injector and method of assembly.
JP6464145B2 (en) 2013-04-05 2019-02-06 ノヴォ・ノルディスク・ヘルス・ケア・アーゲー Growth hormone compound preparation
CA2908198A1 (en) 2013-04-18 2014-10-23 Armo Biosciences, Inc. Methods of using interleukin-10 for treating diseases and disorders
WO2014176373A2 (en) * 2013-04-24 2014-10-30 Armo Biosciences, Inc. Interleukin-10 compositions and uses thereof
EP2988826A1 (en) * 2013-04-26 2016-03-02 Philogen S.p.A. Il4 conjugated to antibodies against extracellular matrix components
US11117975B2 (en) 2013-04-29 2021-09-14 Teva Pharmaceuticals Australia Pty Ltd Anti-CD38 antibodies and fusions to attenuated interferon alpha-2B
BR112015027313A2 (en) 2013-04-29 2017-09-26 Teva Pharmaceuticals Australia Pty Ltd anti-cd38 antibodies and attenuated interferon alfa-2b fusions
EP3010527B1 (en) 2013-06-17 2018-08-08 Armo Biosciences, Inc. Method for assessing protein identity and stability
WO2015000585A1 (en) 2013-07-02 2015-01-08 Walter Sebald Muteins of cytokines of the gamma-chain receptor family conjugated to a non-protein group
ES2658039T3 (en) 2013-07-10 2018-03-08 Sutro Biopharma, Inc. Antibodies comprising multiple site-specific non-natural amino acid residues, methods for their preparation and methods of use
BR112016002845A2 (en) 2013-08-12 2017-09-12 Genentech Inc compositions and methods for treating complement-associated conditions
JP6509867B2 (en) 2013-08-30 2019-05-08 アルモ・バイオサイエンシーズ・インコーポレイテッド Methods of using interleukin-10 to treat diseases and disorders
WO2015036553A1 (en) 2013-09-16 2015-03-19 Novo Nordisk Health Care Ag Thiol functionalized polymers
WO2015054658A1 (en) 2013-10-11 2015-04-16 Sutro Biopharma, Inc. Modified amino acids comprising tetrazine functional groups, methods of preparation, and methods of their use
WO2015061386A1 (en) 2013-10-24 2015-04-30 Amgen Inc. Injector and method of assembly
AU2014340174B2 (en) 2013-10-24 2019-09-12 Amgen Inc. Drug delivery system with temperature-sensitive control
ES2862139T3 (en) 2013-11-11 2021-10-07 Armo Biosciences Inc Procedures for the use of Interleukin 10 for the treatment of diseases and disorders
US9959384B2 (en) 2013-12-10 2018-05-01 Bioniz, Llc Methods of developing selective peptide antagonists
ES2869309T3 (en) 2013-12-13 2021-10-25 Novo Nordisk Healthcare Ag Method for protein conjugation with thioether
WO2015119906A1 (en) 2014-02-05 2015-08-13 Amgen Inc. Drug delivery system with electromagnetic field generator
AU2015253042A1 (en) 2014-05-01 2016-10-20 Genentech, Inc. Anti-factor D antibody variants and uses thereof
UA119352C2 (en) 2014-05-01 2019-06-10 Тева Фармасьютикалз Острейліа Пті Лтд Combination of lenalidomide or pomalidomide and cd38 antibody-attenuated interferon-alpha constructs, and the use thereof
WO2015171777A1 (en) 2014-05-07 2015-11-12 Amgen Inc. Autoinjector with shock reducing elements
CN106573072A (en) 2014-06-02 2017-04-19 阿尔莫生物科技股份有限公司 Methods of lowering serum cholesterol
EP3151883A1 (en) 2014-06-03 2017-04-12 Amgen Inc. Devices and methods for assisting a user of a drug delivery device
EP3197915A4 (en) 2014-09-22 2018-12-19 Intrinsic Lifesciences LLC Humanized anti-hepcidin antibodies and uses thereof
EP3943135A3 (en) 2014-10-14 2022-06-29 Amgen Inc. Drug injection device with visual and audible indicators
KR20170068553A (en) 2014-10-14 2017-06-19 아르모 바이오사이언시스 인코포레이티드 Interleukin-15 compositions and uses thereof
JP6675394B2 (en) 2014-10-22 2020-04-01 アルモ・バイオサイエンシーズ・インコーポレイテッド Use of interleukin-10 for the treatment of diseases and disorders
UY36370A (en) 2014-10-24 2016-04-29 Bristol Myers Squibb Company Una Corporación Del Estado De Delaware MODIFIED FGF-21 POLIPEPTIDES AND ITS USES
WO2016065409A1 (en) * 2014-10-29 2016-05-06 Teva Pharmaceuticals Australia Pty Ltd INTERFERON α2B VARIANTS
EP3689394A1 (en) 2014-12-19 2020-08-05 Amgen Inc. Drug delivery device with live button or user interface field
US10799630B2 (en) 2014-12-19 2020-10-13 Amgen Inc. Drug delivery device with proximity sensor
US10618970B2 (en) 2015-02-03 2020-04-14 Armo Biosciences, Inc. Method of treating cancer with IL-10 and antibodies that induce ADCC
EP3556411B1 (en) 2015-02-17 2021-06-30 Amgen Inc. Drug delivery device with vacuum assisted securement and/or feedback
ES2905870T3 (en) 2015-02-27 2022-04-12 Amgen Inc Drug delivery device having a needle guard mechanism with an adjustable threshold resistance to movement of the needle guard
KR20180020141A (en) 2015-05-28 2018-02-27 아르모 바이오사이언시스 인코포레이티드 PEGylated interleukin-10 for cancer treatment
ES2784603T3 (en) 2015-06-02 2020-09-29 Novo Nordisk As Insulins with recombinant polar extensions
US10874717B2 (en) 2015-07-07 2020-12-29 Burr Oak Therapeutics LLC Pegylated growth hormone antagonists
CN108025040A (en) 2015-08-25 2018-05-11 阿尔莫生物科技股份有限公司 The method that disease and illness are treated using interleukin-10
WO2017039786A1 (en) 2015-09-02 2017-03-09 Amgen Inc. Syringe assembly adapter for a syringe
US11229683B2 (en) 2015-09-18 2022-01-25 Bolder Biotechnology, Inc. Hematopoietic growth factor proteins and analogs thereof and angiotensin converting enzyme inhibitors for treatment of radiation exposure
MA43348A (en) 2015-10-01 2018-08-08 Novo Nordisk As PROTEIN CONJUGATES
PT3359556T (en) 2015-10-09 2021-08-30 Bioniz Llc Modulating gamma - c -cytokine activity
WO2017075252A1 (en) 2015-10-30 2017-05-04 Genentech, Inc. Anti-factor d antibody variant conjugates and uses thereof
WO2017075173A2 (en) 2015-10-30 2017-05-04 Genentech, Inc. Anti-factor d antibodies and conjugates
KR20180077271A (en) 2015-11-03 2018-07-06 암브룩스, 인코포레이티드 Anti-CD3-Folate conjugates and uses thereof
WO2017100501A1 (en) 2015-12-09 2017-06-15 Amgen Inc. Auto-injector with signaling cap
US11154661B2 (en) 2016-01-06 2021-10-26 Amgen Inc. Auto-injector with signaling electronics
WO2017160799A1 (en) 2016-03-15 2017-09-21 Amgen Inc. Reducing probability of glass breakage in drug delivery devices
WO2017189432A1 (en) 2016-04-26 2017-11-02 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same
US11541168B2 (en) 2016-04-29 2023-01-03 Amgen Inc. Drug delivery device with messaging label
US11389588B2 (en) 2016-05-02 2022-07-19 Amgen Inc. Syringe adapter and guide for filling an on-body injector
ES2959783T3 (en) 2016-05-13 2024-02-28 Amgen Inc Vial Protective Cover Assembly
EP3458988B1 (en) 2016-05-16 2023-10-18 Amgen Inc. Data encryption in medical devices with limited computational capability
EP3465124A1 (en) 2016-06-03 2019-04-10 Amgen Inc. Impact testing apparatuses and methods for drug delivery devices
TW201808987A (en) * 2016-06-08 2018-03-16 健生生物科技公司 GM-CSF variants and methods of use
EP3478342A1 (en) 2016-07-01 2019-05-08 Amgen Inc. Drug delivery device having minimized risk of component fracture upon impact events
WO2018034784A1 (en) 2016-08-17 2018-02-22 Amgen Inc. Drug delivery device with placement detection
EP3532127A1 (en) 2016-10-25 2019-09-04 Amgen Inc. On-body injector
WO2018136398A1 (en) 2017-01-17 2018-07-26 Amgen Inc. Injection devices and related methods of use and assembly
AU2018219283B2 (en) 2017-02-08 2022-05-19 Bristol-Myers Squibb Company Modified relaxin polypeptides comprising a pharmacokinetic enhancer and uses thereof
MX2019009625A (en) 2017-02-17 2019-10-09 Amgen Inc Drug delivery device with sterile fluid flowpath and related method of assembly.
CA3052204A1 (en) 2017-02-17 2018-08-23 Amgen Inc. Insertion mechanism for drug delivery device
EP3592403A1 (en) 2017-03-06 2020-01-15 Amgen Inc. Drug delivery device with activation prevention feature
JP2020509839A (en) 2017-03-07 2020-04-02 アムジエン・インコーポレーテツド Needle insertion due to overpressure
MX2019010671A (en) 2017-03-09 2019-10-21 Amgen Inc Insertion mechanism for drug delivery device.
CN110446499A (en) 2017-03-20 2019-11-12 豪夫迈·罗氏有限公司 A kind of method of external glycosyl engineering erythropoiesis stimulating protein
AU2018245995B2 (en) 2017-03-28 2024-03-07 Amgen Inc. Plunger rod and syringe assembly system and method
JP2020513019A (en) 2017-04-05 2020-04-30 ノヴォ ノルディスク アー/エス Oligomer-extended insulin-Fc conjugate
CN110709121B (en) 2017-06-08 2022-06-24 安进公司 Torque-driven drug delivery device
US11590294B2 (en) 2017-06-08 2023-02-28 Amgen Inc. Syringe assembly for a drug delivery device and method of assembly
JP7195276B2 (en) 2017-06-22 2022-12-23 アムジエン・インコーポレーテツド Collision/shock reduction due to device activation
EP3641861A1 (en) 2017-06-23 2020-04-29 Amgen Inc. Electronic drug delivery device comprising a cap activated by a switch assembly
MA49562A (en) 2017-07-14 2020-05-20 Amgen Inc NEEDLE INSERTION-RETRACTION SYSTEM FEATURING A DOUBLE-TORSION SPRING SYSTEM
WO2019018169A1 (en) 2017-07-21 2019-01-24 Amgen Inc. Gas permeable sealing member for drug container and methods of assembly
EP3658206A1 (en) 2017-07-25 2020-06-03 Amgen Inc. Drug delivery device with container access system and related method of assembly
JP2020528296A (en) 2017-07-25 2020-09-24 アムジエン・インコーポレーテツド Drug delivery device with gear module and related assembly method
EP3664863A2 (en) 2017-08-09 2020-06-17 Amgen Inc. Hydraulic-pneumatic pressurized chamber drug delivery system
WO2019036181A1 (en) 2017-08-18 2019-02-21 Amgen Inc. Wearable injector with sterile adhesive patch
US11103636B2 (en) 2017-08-22 2021-08-31 Amgen Inc. Needle insertion mechanism for drug delivery device
CN111050786A (en) 2017-09-08 2020-04-21 百时美施贵宝公司 Modified fibroblast growth factor 21(FGF-21) for use in a method of treating non-alcoholic steatohepatitis (NASH)
ES2939292T3 (en) 2017-10-04 2023-04-20 Amgen Inc Flow adapter for drug delivery device
US11813426B2 (en) 2017-10-06 2023-11-14 Amgen Inc. Drug delivery device including seal member for needle of syringe
EP3694578A1 (en) 2017-10-09 2020-08-19 Amgen Inc. Drug delivery device with drive assembly and related method of assembly
MA50528A (en) 2017-11-03 2020-09-09 Amgen Inc SYSTEMS AND APPROACHES TO STERILIZE A DRUG DELIVERY DEVICE
WO2019089178A1 (en) 2017-11-06 2019-05-09 Amgen Inc. Drug delivery device with placement and flow sensing
US20200338271A1 (en) 2017-11-06 2020-10-29 Amgen Inc. Fill-finish assemblies and related methods
MA50557A (en) 2017-11-10 2020-09-16 Amgen Inc PISTONS FOR DRUG DELIVERY DEVICES
CA3079540A1 (en) 2017-11-16 2019-05-23 Amgen Inc. Door latch mechanism for drug delivery device
MX2020005066A (en) 2017-11-16 2020-08-20 Amgen Inc Autoinjector with stall and end point detection.
AU2018394189B2 (en) * 2017-12-27 2023-12-21 Kyowa Kirin Co., Ltd. IL-2 variant
WO2019130344A1 (en) * 2017-12-27 2019-07-04 Council Of Scientific And Industrial Research A polypeptide exhibiting granulocyte-colony stimulating factor activity
US10835685B2 (en) 2018-05-30 2020-11-17 Amgen Inc. Thermal spring release mechanism for a drug delivery device
US11083840B2 (en) 2018-06-01 2021-08-10 Amgen Inc. Modular fluid path assemblies for drug delivery devices
CN114903978A (en) 2018-07-03 2022-08-16 百时美施贵宝公司 FGF-21 formulations
CN112469454B (en) 2018-07-24 2024-01-26 安进公司 Delivery device for administering a drug
WO2020023220A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Hybrid drug delivery devices with tacky skin attachment portion and related method of preparation
US20210128844A1 (en) 2018-07-24 2021-05-06 Amgen Inc. Delivery devices for administering drugs
WO2020023336A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Hybrid drug delivery devices with grip portion
US20210228797A1 (en) 2018-07-31 2021-07-29 Amgen Inc. Fluid path assembly for a drug delivery device
SG11202101875VA (en) 2018-08-28 2021-03-30 Ambrx Inc Anti-cd3 antibody folate bioconjugates and their uses
US20220056093A1 (en) 2018-09-11 2022-02-24 Ambrx, Inc. Interleukin-2 polypeptide conjugates and their uses
EP3856284A1 (en) 2018-09-24 2021-08-04 Amgen Inc. Interventional dosing systems and methods
EP3856283A1 (en) 2018-09-28 2021-08-04 Amgen Inc. Muscle wire escapement activation assembly for a drug delivery device
TW202027806A (en) 2018-10-02 2020-08-01 美商安進公司 Injection systems for drug delivery with internal force transmission
TWI824026B (en) 2018-10-05 2023-12-01 美商安進公司 Drug delivery device having dose indicator
AR116704A1 (en) 2018-10-15 2021-06-02 Amgen Inc DRUG ADMINISTRATION DEVICE WITH CUSHIONING MECHANISM
SG11202101824VA (en) 2018-10-15 2021-03-30 Amgen Inc Platform assembly process for drug delivery device
CN113366015A (en) 2018-10-19 2021-09-07 Ambrx公司 Interleukin-10 polypeptide conjugates, dimers thereof and uses thereof
MA54066A (en) 2018-11-01 2022-02-09 Amgen Inc PARTIAL NEEDLE RETRACTION DRUG DELIVERY DEVICES
MA54048A (en) 2018-11-01 2022-02-09 Amgen Inc DRUG DELIVERY DEVICES WITH PARTIAL RETRACTION OF DRUG DELIVERY BODY
WO2020091981A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
EP3876996A1 (en) 2018-11-05 2021-09-15 Bristol-Myers Squibb Company Method for purifying pegylated protein
CN111378026A (en) * 2018-12-27 2020-07-07 天津键凯科技有限公司 Method for preparing PEG (polyethylene glycol) biological molecules with controllable binding sites
US20220226488A1 (en) 2019-02-12 2022-07-21 Ambrx, Inc. Compositions containing, methods and uses of antibody-tlr agonist conjugates
AU2020258384A1 (en) 2019-04-15 2021-11-04 Qwixel Therapeutics Llc Fusion protein composition(s) comprising targeted masked type I interferons (IFNA and IFNB) and an antibody against tumor antigen, for use in the treatment of cancer
WO2020219482A1 (en) 2019-04-24 2020-10-29 Amgen Inc. Syringe sterilization verification assemblies and methods
EP4017560A2 (en) 2019-08-23 2022-06-29 Amgen, Inc Drug delivery device with configurable needle shield engagement components and related methods
WO2021173889A1 (en) 2020-02-26 2021-09-02 Ambrx, Inc. Uses of anti-cd3 antibody folate bioconjugates
MX2022011163A (en) 2020-03-11 2022-10-18 Ambrx Inc Interleukin-2 polypeptide conjugates and methods of use thereof.
JP2023519213A (en) 2020-03-24 2023-05-10 ジェネンテック, インコーポレイテッド TIE2 binding agents and methods of use
US20220033804A1 (en) * 2020-07-30 2022-02-03 Georgia Tech Research Corporation Methods and Compositions for Enhancement of Stem Cell-based Immunomodulation and Tissue Repair
CA3190606A1 (en) 2020-08-20 2022-02-24 Ambrx, Inc. Antibody-tlr agonist conjugates, methods and uses thereof
WO2022159414A1 (en) 2021-01-22 2022-07-28 University Of Rochester Erythropoietin for gastroinfestinal dysfunction
CA3213805A1 (en) 2021-04-03 2022-10-06 Feng Tian Anti-her2 antibody-drug conjugates and uses thereof
IL307418A (en) 2021-05-21 2023-12-01 Amgen Inc Method of optimizing a filling recipe for a drug container
CN114920819A (en) * 2022-06-16 2022-08-19 修正生物医药(杭州)研究院有限公司 Recombinant human growth hormone mutant, polyethylene glycol derivative thereof, preparation method of polyethylene glycol derivative and pharmaceutical application of polyethylene glycol derivative
WO2024026224A1 (en) 2022-07-29 2024-02-01 University Of Rochester Methods for improving muscle mass, strength, or function with a combination of testosterone and growth hormone

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4588585A (en) * 1982-10-19 1986-05-13 Cetus Corporation Human recombinant cysteine depleted interferon-β muteins
US4810643A (en) * 1985-08-23 1989-03-07 Kirin- Amgen Inc. Production of pluripotent granulocyte colony-stimulating factor
US4904584A (en) * 1987-12-23 1990-02-27 Genetics Institute, Inc. Site-specific homogeneous modification of polypeptides
US5166322A (en) * 1989-04-21 1992-11-24 Genetics Institute Cysteine added variants of interleukin-3 and chemical modifications thereof
US5208158A (en) * 1990-04-19 1993-05-04 Novo Nordisk A/S Oxidation stable detergent enzymes
US5214132A (en) * 1986-12-23 1993-05-25 Kyowa Hakko Kogyo Co., Ltd. Polypeptide derivatives of human granulocyte colony stimulating factor
US5681720A (en) * 1986-12-23 1997-10-28 Kyowa Hakko Co., Ltd. DNA encoding human granulocyte colony stimulating factor plasmids and host cells comprising same, and methods of expressing the encoded polypeptide
US5766897A (en) * 1990-06-21 1998-06-16 Incyte Pharmaceuticals, Inc. Cysteine-pegylated proteins
US5824784A (en) * 1994-10-12 1998-10-20 Amgen Inc. N-terminally chemically modified protein compositions and methods
US5824778A (en) * 1988-12-22 1998-10-20 Kirin-Amgen, Inc. Chemically-modified G-CSF
US5849535A (en) * 1995-09-21 1998-12-15 Genentech, Inc. Human growth hormone variants
US6632426B2 (en) * 1993-01-28 2003-10-14 Amgen Inc. G-CSF analog compositions and methods

Family Cites Families (88)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4678751A (en) * 1981-09-25 1987-07-07 Genentech, Inc. Hybrid human leukocyte interferons
US5582824A (en) 1981-10-19 1996-12-10 Genentech, Inc. Recombinant DES-CYS-TYR-CYS human immune interferon
AU561343B2 (en) 1981-10-19 1987-05-07 Genentech Inc. Human immune interferon by recombinant dna
US5096705A (en) 1981-10-19 1992-03-17 Genentech, Inc. Human immune interferon
US4921699A (en) * 1983-06-01 1990-05-01 Hoffman-La Roche Inc. Polypeptides having interferon activity
GB8317880D0 (en) * 1983-07-01 1983-08-03 Searle & Co Structure and synthesis of interferons
GB8334102D0 (en) * 1983-12-21 1984-02-01 Searle & Co Interferons with cysteine pattern
US4703008A (en) 1983-12-13 1987-10-27 Kiren-Amgen, Inc. DNA sequences encoding erythropoietin
KR850004274A (en) 1983-12-13 1985-07-11 원본미기재 Method for preparing erythropoietin
MX9203641A (en) 1983-12-16 1992-07-01 Genentech Inc RECOMBINANT GAMMA INTERFERONS THAT HAVE IMPROVED STABILITY AND BIOTECHNOLOGICAL METHODS FOR THEIR OBTAINING.
US4855238A (en) 1983-12-16 1989-08-08 Genentech, Inc. Recombinant gamma interferons having enhanced stability and methods therefor
AU594014B2 (en) * 1984-03-21 1990-03-01 Research Corporation Technologies, Inc. Recombinant DNA molecules
US4636463A (en) * 1984-04-05 1987-01-13 Scripps Clinic And Research Foundation Antibodies to human interleukin-2 induced by synthetic polypeptides
US5908763A (en) * 1984-07-06 1999-06-01 Novartis Corporation DNA encoding GM-CSF and a method of producing GM-CSF protein
RU2130493C1 (en) * 1984-07-06 1999-05-20 Новартис Аг Method of preparing protein exhibiting gm-csf activity of pimates
UA29377C2 (en) * 1984-09-19 2000-11-15 Новартіс Аг Method for preparing protein with activity of granulocyte macrophage-colony stimulating factor (gm-csf) of primates
US4572798A (en) 1984-12-06 1986-02-25 Cetus Corporation Method for promoting disulfide bond formation in recombinant proteins
DK151585D0 (en) * 1985-04-03 1985-04-03 Nordisk Gentofte DNA sequence
US5206344A (en) 1985-06-26 1993-04-27 Cetus Oncology Corporation Interleukin-2 muteins and polymer conjugation thereof
US5391485A (en) * 1985-08-06 1995-02-21 Immunex Corporation DNAs encoding analog GM-CSF molecules displaying resistance to proteases which cleave at adjacent dibasic residues
DE3540076A1 (en) 1985-11-12 1987-05-14 Boehringer Mannheim Gmbh METHOD FOR STABILIZING CREATINE KINASE
DK203187A (en) 1986-04-22 1987-10-23 Immunex Corp HUMAN G-CSF PROTEIN EXPRESSION
JPH01132598A (en) 1987-10-14 1989-05-25 Pitman Moore Inc Method for promoting production of intramolecular disulfide bond in recominant protein contained in modifier solution
AU3532289A (en) 1988-05-06 1989-11-29 Genentech Inc. Novel bovine granulocyte-macrophage colony stimulating factor variant
ATE198353T1 (en) 1988-08-24 2001-01-15 American Cyanamid Co STABILIZATION OF SOMATOTROPINES THROUGH MODIFICATION OF CYSTEIN RESIDUE THROUGH SITE-SPECIFIC MUtagenesis OR CHEMICAL DERIVATIZATION
US5223407A (en) * 1988-08-31 1993-06-29 Allelix Inc. Excretion of heterologous proteins from e. coli
US6780613B1 (en) * 1988-10-28 2004-08-24 Genentech, Inc. Growth hormone variants
JP2517100B2 (en) 1989-03-08 1996-07-24 協和醗酵工業株式会社 Purification method of protein having human granulocyte colony-stimulating factor activity
US5130418A (en) 1989-05-02 1992-07-14 California Biotechnology Inc. Method to stabilize basic fibroblast growth factor
JPH04164098A (en) 1990-03-07 1992-06-09 Kirin Amgen Inc Chemically modified, granular spherical colony stimulus factor derivative
US5235043A (en) 1990-04-06 1993-08-10 Synergen, Inc. Production of biologically active, recombinant members of the ngf/bdnf family of neurotrophic proteins
US6552170B1 (en) * 1990-04-06 2003-04-22 Amgen Inc. PEGylation reagents and compounds formed therewith
ATE163431T1 (en) 1990-05-04 1998-03-15 American Cyanamid Co STABILIZATION OF SOMATOTROPIN THROUGH MODIFICATION OF CYSTEINE RESIDUE
DE4113750A1 (en) 1991-04-26 1992-10-29 Boehringer Mannheim Gmbh IMPROVEMENT OF RENATURATION IN THE SECRETION OF DISULFID-BRIDGED PROTEINS
WO1993000109A1 (en) 1991-06-28 1993-01-07 Genentech, Inc. Method of stimulating immune response using growth hormone
US6171824B1 (en) 1991-08-30 2001-01-09 Fred Hutchinson Cancer Research Center Hybrid cytokines
JP3951062B2 (en) 1991-09-19 2007-08-01 ジェネンテック・インコーポレーテッド Expression of antibody fragments with cysteine present at least as a free thiol in E. coli for the production of bifunctional F (ab ') 2 antibodies
EP0650497A1 (en) 1992-07-02 1995-05-03 Mallinckrodt Veterinary, Inc. A process for recovering a recombinant protein, in biologically active form, from a solution containing inactive protein
US5382657A (en) * 1992-08-26 1995-01-17 Hoffmann-La Roche Inc. Peg-interferon conjugates
EP0672145B1 (en) 1992-11-24 2003-04-23 G.D. Searle & Co. Interleukin-3 (il-3) multiple mutation polypeptides
JPH08506095A (en) * 1992-11-25 1996-07-02 アムジェン ボールダー インコーポレイテッド Modified insulin-like growth factor
US5441734A (en) * 1993-02-25 1995-08-15 Schering Corporation Metal-interferon-alpha crystals
AU6626794A (en) * 1993-04-07 1994-10-24 Amgen Boulder Inc. Methods of using insulin-like growth factor binding proteins
EP0626448A3 (en) * 1993-05-26 1998-01-14 BOEHRINGER INGELHEIM INTERNATIONAL GmbH Process for preparing and purifying alpha-interferon
JPH09504174A (en) * 1993-10-29 1997-04-28 インサイト ファーマシューティカルズ,インク. Chimeric protein containing protease nexin 1 mutant
US5951974A (en) * 1993-11-10 1999-09-14 Enzon, Inc. Interferon polymer conjugates
US6207640B1 (en) 1994-04-07 2001-03-27 Genentech, Inc. Treatment of partial growth hormone insensitivity syndrome
AU2601895A (en) 1994-05-24 1995-12-18 Amgen Boulder Inc. Modified insulin-like growth factors
ITFI940106A1 (en) * 1994-05-27 1995-11-27 Menarini Ricerche Sud Spa HYBRID MOLECULE OF GM-CSF-L-EPO OR EPO-L-GM-CSF FORMULA FOR ERYTHROPOIETIC STIMULATION
CA2172273A1 (en) 1994-07-29 1996-02-15 Geert Maertens Purified hepatitis c virus envelope proteins for diagnostic and therapeutic use
PT785948E (en) 1994-10-13 2003-09-30 Amgen Inc ANALYSIS OF THE KERATINOCYTE GROWTH FACTOR
AU5386996A (en) 1995-04-05 1996-10-23 Picower Institute For Medical Research, The Agents for binding to advanced glycosylation endproducts, an d methods of their use
WO1996036362A1 (en) 1995-05-16 1996-11-21 Prizm Pharmaceuticals, Inc. Compositions containing nucleic acids and ligands for therapeutic treatment
DE19535853C2 (en) 1995-09-18 1999-04-01 Fraunhofer Ges Forschung Variants of recombinant human interferon gamma, process for their preparation and their use
WO1997011370A1 (en) 1995-09-22 1997-03-27 Immunomedics, Inc. Recombinant proteins having multiple disulfide bonds and thiol-substituted conjugates thereof
JP2000507917A (en) * 1995-11-02 2000-06-27 シェーリング コーポレイション Continuous low-dose cytokine infusion therapy
US5908621A (en) * 1995-11-02 1999-06-01 Schering Corporation Polyethylene glycol modified interferon therapy
GB9625640D0 (en) 1996-12-10 1997-01-29 Celltech Therapeutics Ltd Biological products
BR9606270A (en) * 1996-12-18 1998-09-22 Univ Minas Gerais Process for the production of recombinant human beta-cis interferon protein and recombinant human beta-cis interferon protein
CA2615918C (en) 1997-02-21 2013-11-26 Genentech, Inc. Antibody fragment-polymer conjugates and humanized anti-il-8 monoclonal antibodies
DE19717864C2 (en) * 1997-04-23 2001-05-17 Fraunhofer Ges Forschung Human recombinant interferon beta, process for its preparation and its use
US7153943B2 (en) 1997-07-14 2006-12-26 Bolder Biotechnology, Inc. Derivatives of growth hormone and related proteins, and methods of use thereof
US6753165B1 (en) 1999-01-14 2004-06-22 Bolder Biotechnology, Inc. Methods for making proteins containing free cysteine residues
EP1881005B1 (en) 1997-07-14 2013-04-03 Bolder Biotechnology, Inc. Derivatives of G-CSF and related proteins
US7270809B2 (en) * 1997-07-14 2007-09-18 Bolder Biotechnology, Inc. Cysteine variants of alpha interferon-2
US20080076706A1 (en) * 1997-07-14 2008-03-27 Bolder Biotechnology, Inc. Derivatives of Growth Hormone and Related Proteins, and Methods of Use Thereof
US7495087B2 (en) 1997-07-14 2009-02-24 Bolder Biotechnology, Inc. Cysteine muteins in the C-D loop of human interleukin-11
US7306931B2 (en) 2000-05-16 2007-12-11 Bolder Biotechnology, Inc. Method for refolding proteins containing free cysteine residues
US6472373B1 (en) * 1997-09-21 2002-10-29 Schering Corporation Combination therapy for eradicating detectable HCV-RNA in antiviral treatment naive patients having chronic hepatitis C infection
US6756491B2 (en) * 1998-01-09 2004-06-29 The Salk Institute For Biological Studies Steroid-activated nuclear receptors and uses therefor
US6180096B1 (en) * 1998-03-26 2001-01-30 Schering Corporation Formulations for protection of peg-interferon alpha conjugates
US8288126B2 (en) * 1999-01-14 2012-10-16 Bolder Biotechnology, Inc. Methods for making proteins containing free cysteine residues
BR0008759B1 (en) 1999-01-14 2014-03-11 Bolder Biotechnology Inc Methods for the production of protein containing free cysteine residues
US6362162B1 (en) * 1999-04-08 2002-03-26 Schering Corporation CML Therapy
US6514729B1 (en) 1999-05-12 2003-02-04 Xencor, Inc. Recombinant interferon-beta muteins
US6531122B1 (en) * 1999-08-27 2003-03-11 Maxygen Aps Interferon-β variants and conjugates
JP4617533B2 (en) * 2000-03-14 2011-01-26 ソニー株式会社 Information providing apparatus and method, information processing apparatus and method, and program storage medium
WO2002044717A1 (en) 2000-12-01 2002-06-06 Cornell Research Foundation Animal model for flaviviridae infection
US7038015B2 (en) 2001-04-06 2006-05-02 Maxygen Holdings, Ltd. Interferon gamma polypeptide variants
US20030233694A1 (en) * 2002-06-25 2003-12-25 Michael Wescombe-Down Protective swimsuit incorporating an electrical wiring system
AU2003270341A1 (en) 2002-09-05 2004-03-29 The General Hospital Corporation Modified asialo-interferons and uses thereof
JP4507099B2 (en) 2004-07-09 2010-07-21 エルピーダメモリ株式会社 Semiconductor device module
MX2007002441A (en) 2004-08-31 2007-05-04 Pharmacia & Upjohn Co Llc Glycerol branched polyethylene glycol human growth hormone conjugates, process for their preparation, and methods of use thereof.
BRPI0713963A2 (en) 2006-07-07 2012-11-27 Novo Nordisk Healthcare Ag protein conjugates and methods for their preparation
CN105963710A (en) 2009-08-06 2016-09-28 诺沃—诺迪斯克保健股份有限公司 Growth hormones with prolonged in-vivo efficacy
JP5268983B2 (en) 2010-04-05 2013-08-21 株式会社エヌ・ティ・ティ・ドコモ COMMUNICATION CONTROL METHOD, MOBILE STATION DEVICE, AND BASE STATION DEVICE
AU2014286998B2 (en) 2013-07-11 2020-01-30 The Scripps Research Institute Coiled coil immunoglobulin fusion proteins and compositions thereof
US9506540B1 (en) 2015-12-29 2016-11-29 Honda Motor Co., Ltd. Pedal apparatus

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4588585A (en) * 1982-10-19 1986-05-13 Cetus Corporation Human recombinant cysteine depleted interferon-β muteins
US4810643A (en) * 1985-08-23 1989-03-07 Kirin- Amgen Inc. Production of pluripotent granulocyte colony-stimulating factor
US5214132A (en) * 1986-12-23 1993-05-25 Kyowa Hakko Kogyo Co., Ltd. Polypeptide derivatives of human granulocyte colony stimulating factor
US5681720A (en) * 1986-12-23 1997-10-28 Kyowa Hakko Co., Ltd. DNA encoding human granulocyte colony stimulating factor plasmids and host cells comprising same, and methods of expressing the encoded polypeptide
US4904584A (en) * 1987-12-23 1990-02-27 Genetics Institute, Inc. Site-specific homogeneous modification of polypeptides
US5824778A (en) * 1988-12-22 1998-10-20 Kirin-Amgen, Inc. Chemically-modified G-CSF
US5166322A (en) * 1989-04-21 1992-11-24 Genetics Institute Cysteine added variants of interleukin-3 and chemical modifications thereof
US5208158A (en) * 1990-04-19 1993-05-04 Novo Nordisk A/S Oxidation stable detergent enzymes
US5766897A (en) * 1990-06-21 1998-06-16 Incyte Pharmaceuticals, Inc. Cysteine-pegylated proteins
US6632426B2 (en) * 1993-01-28 2003-10-14 Amgen Inc. G-CSF analog compositions and methods
US5824784A (en) * 1994-10-12 1998-10-20 Amgen Inc. N-terminally chemically modified protein compositions and methods
US5849535A (en) * 1995-09-21 1998-12-15 Genentech, Inc. Human growth hormone variants

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080076706A1 (en) * 1997-07-14 2008-03-27 Bolder Biotechnology, Inc. Derivatives of Growth Hormone and Related Proteins, and Methods of Use Thereof
US20080075672A1 (en) * 2002-09-09 2008-03-27 Rene Gantier Rational evolution of cytokines for higher stability, the cytokines and encoding nucleic acid molecules
WO2009139784A1 (en) * 2008-05-13 2009-11-19 Nora Therapeutics, Inc. Human g-csf analogs and methods of making and using thereof
US20110171168A1 (en) * 2008-05-13 2011-07-14 Nora Therapeutics, Inc. Human g-csf analogs and methods of making and using thereof

Also Published As

Publication number Publication date
ATE375363T1 (en) 2007-10-15
US20050214254A1 (en) 2005-09-29
US20170369547A1 (en) 2017-12-28
US7148333B2 (en) 2006-12-12
US20050227330A1 (en) 2005-10-13
US10329337B2 (en) 2019-06-25
EP1012184A4 (en) 2003-03-26
EP1012184B1 (en) 2007-10-10
AU751898B2 (en) 2002-08-29
EP1012184A1 (en) 2000-06-28
JP2009096810A (en) 2009-05-07
DE69838552D1 (en) 2007-11-22
US7253267B2 (en) 2007-08-07
US20040230040A1 (en) 2004-11-18
US8618256B2 (en) 2013-12-31
US7309781B2 (en) 2007-12-18
NZ502375A (en) 2001-11-30
US6608183B1 (en) 2003-08-19
US7314921B2 (en) 2008-01-01
US7959909B2 (en) 2011-06-14
US7345149B2 (en) 2008-03-18
ES2297889T3 (en) 2008-05-01
US20140163204A1 (en) 2014-06-12
IL133974A (en) 2010-05-17
DE69838552T2 (en) 2008-05-21
CA2296770A1 (en) 1999-01-28
BR9812267B1 (en) 2013-11-26
US7964184B2 (en) 2011-06-21
US7732572B2 (en) 2010-06-08
US20040175356A1 (en) 2004-09-09
US20040175800A1 (en) 2004-09-09
US20080317713A1 (en) 2008-12-25
US20050107591A1 (en) 2005-05-19
US7345154B2 (en) 2008-03-18
IL200623A (en) 2012-02-29
US20040214287A1 (en) 2004-10-28
US20090060863A1 (en) 2009-03-05
US20030162949A1 (en) 2003-08-28
US20110250169A1 (en) 2011-10-13
US7214779B2 (en) 2007-05-08
WO1999003887A1 (en) 1999-01-28
US20030166865A1 (en) 2003-09-04
HK1116497A1 (en) 2008-12-24
HK1028617A1 (en) 2001-02-23
US20050096461A1 (en) 2005-05-05
US7232885B2 (en) 2007-06-19
EP1881005A1 (en) 2008-01-23
AU8300098A (en) 1999-02-10
US20080219950A1 (en) 2008-09-11
EP1881005B1 (en) 2013-04-03
IL133974A0 (en) 2001-04-30
US7795396B2 (en) 2010-09-14
JP2001510033A (en) 2001-07-31
BR9812267A (en) 2001-12-18
CN1269805A (en) 2000-10-11
IL200623A0 (en) 2011-07-31

Similar Documents

Publication Publication Date Title
US7309781B2 (en) Cysteine variants of granulocyte colony-stimulating factor
US8148500B2 (en) Method of preparing cysteine mutants of human recombinant GM-CSF
AU2008203051B2 (en) Derivatives of growth hormone and related proteins
AU2002306305C1 (en) Derivatives of growth hormone and related proteins
AU2006203197B2 (en) Derivatives of growth hormone and related proteins

Legal Events

Date Code Title Description
STCF Information on status: patent grant

Free format text: PATENTED CASE

CC Certificate of correction
FPAY Fee payment

Year of fee payment: 4

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:BOLDER BIOTECHNOLOGY, INC.;REEL/FRAME:033889/0200

Effective date: 20140929

FPAY Fee payment

Year of fee payment: 8

CC Certificate of correction
FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20191218